The incidence peak of cancer is around the 6th decade of life. Therefore, it isn’t important in the evolution or in shaping the immune system → mainly occurs after reproductive age.In the beginning of the 20th century, a number of scientists proposed a theory of immune surveillance, which later was further confirmed:If a mouse is injected with a carcinogen, a tumor starts developingIf the tumor is injected into another mouse, the tumor continues developingIf the tumor is first subjected to radiation and vaccinated into a mouse, the tumor stops growingThis shows that the immune system plays an important role in tumor growth. The presence of certain immune cells correlates with both improved and worse tumor behavior and prognosis. T-cells are associated with both a good and bad prognosis, depending on the type:Presence of CD8 T-cells correlates with a good prognosisPresence of tertiary lymphoid structures are a good signPresence of regulatory T-cells are associated with a worse prognosis → immune suppressive role Macrophages are often associated with immunosuppressive activity. They can produce cytokines like IL-10 or TGF-β, which counteract immune activity. There are 2 types of macrophages:M1: pro-inflammatory → better prognosisM2: immunosuppressive → worse prognosisThe majority of macrophages in tumors are M2 → make a bad prognosis. When a cell is targeted by carcinogens such as UV-radiation or cytotoxic drugs, the following happens:Damage response pathways are activated, such as:ATMP53When these proteins are activated, they induce the expression of a number of stress molecules which go to the surface of cellsThe proteins are recognized by NK-cells or CD8 T-cellsThe target...


Access options

      How do you get full online access and services on JoHo WorldSupporter.org?

      1 - Go to www JoHo.org, and join JoHo WorldSupporter by choosing a membership + online access
       
      2 - Return to WorldSupporter.org and create an account with the same email address
       
      3 - State your JoHo WorldSupporter Membership during the creation of your account, and you can start using the services
      • You have online access to all free + all exclusive summaries and study notes on WorldSupporter.org and JoHo.org
      • You can use all services on JoHo WorldSupporter.org (EN/NL)
      • You can make use of the tools for work abroad, long journeys, voluntary work, internships and study abroad on JoHo.org (Dutch service)
      Already an account?
      • If you already have a WorldSupporter account than you can change your account status from 'I am not a JoHo WorldSupporter Member' into 'I am a JoHo WorldSupporter Member with full online access
      • Please note: here too you must have used the same email address.
      Are you having trouble logging in or are you having problems logging in?

      Toegangsopties (NL)

      Hoe krijg je volledige toegang en online services op JoHo WorldSupporter.org?

      1 - Ga naar www JoHo.org, en sluit je aan bij JoHo WorldSupporter door een membership met online toegang te kiezen
      2 - Ga terug naar WorldSupporter.org, en maak een account aan met hetzelfde e-mailadres
      3 - Geef bij het account aanmaken je JoHo WorldSupporter membership aan, en je kunt je services direct gebruiken
      • Je hebt nu online toegang tot alle gratis en alle exclusieve samenvattingen en studiehulp op WorldSupporter.org en JoHo.org
      • Je kunt gebruik maken van alle diensten op JoHo WorldSupporter.org (EN/NL)
      • Op JoHo.org kun je gebruik maken van de tools voor werken in het buitenland, verre reizen, vrijwilligerswerk, stages en studeren in het buitenland
      Heb je al een WorldSupporter account?
      • Wanneer je al eerder een WorldSupporter account hebt aangemaakt dan kan je, nadat je bent aangesloten bij JoHo via je 'membership + online access ook je status op WorldSupporter.org aanpassen
      • Je kunt je status aanpassen van 'I am not a JoHo WorldSupporter Member' naar 'I am a JoHo WorldSupporter Member with 'full online access'.
      • Let op: ook hier moet je dan wel hetzelfde email adres gebruikt hebben
      Kom je er niet helemaal uit of heb je problemen met inloggen?

      Join JoHo WorldSupporter!

      What can you choose from?

      JoHo WorldSupporter membership (= from €5 per calendar year):
      • To support the JoHo WorldSupporter and Smokey projects and to contribute to all activities in the field of international cooperation and talent development
      • To use the basic features of JoHo WorldSupporter.org
      JoHo WorldSupporter membership + online access (= from €10 per calendar year):
      • To support the JoHo WorldSupporter and Smokey projects and to contribute to all activities in the field of international cooperation and talent development
      • To use full services on JoHo WorldSupporter.org (EN/NL)
      • For access to the online book summaries and study notes on JoHo.org and Worldsupporter.org
      • To make use of the tools for work abroad, long journeys, voluntary work, internships and study abroad on JoHo.org (NL service)

      Sluit je aan bij JoHo WorldSupporter!  (NL)

      Waar kan je uit kiezen?

      JoHo membership zonder extra services (donateurschap) = €5 per kalenderjaar
      • Voor steun aan de JoHo WorldSupporter en Smokey projecten en een bijdrage aan alle activiteiten op het gebied van internationale samenwerking en talentontwikkeling
      • Voor gebruik van de basisfuncties van JoHo WorldSupporter.org
      • Voor het gebruik van de kortingen en voordelen bij partners
      • Voor gebruik van de voordelen bij verzekeringen en reisverzekeringen zonder assurantiebelasting
      JoHo membership met extra services (abonnee services):  Online toegang Only= €10 per kalenderjaar
      • Voor volledige online toegang en gebruik van alle online boeksamenvattingen en studietools op WorldSupporter.org en JoHo.org
      • voor online toegang tot de tools en services voor werk in het buitenland, lange reizen, vrijwilligerswerk, stages en studie in het buitenland
      • voor online toegang tot de tools en services voor emigratie of lang verblijf in het buitenland
      • voor online toegang tot de tools en services voor competentieverbetering en kwaliteitenonderzoek
      • Voor extra steun aan JoHo, WorldSupporter en Smokey projecten

      Meld je aan, wordt donateur en maak gebruik van de services

      Check page access:
      JoHo members
      Check more or recent content:

      Mechanisms of Disease 2 2020/2021 UL

      Mechanisms of Disease 2 HC2: Cancer genetics

      Mechanisms of Disease 2 HC2: Cancer genetics

      HC2: Cancer genetics

      Multistage evolution of cancer

      A normal epithelial cell has to go through several stages to be transformed into a metastasizing tumor. There is a progressive development from normal to neoplastic tissue, with many non-malignant intermediate stages.

      Clonal expansion model:

      Cancer development is driven by the accumulation of mutations:

      1. An initiating mutation occurs in a normal cell → gives the cell a growth advantage over his neighboring cells
      2. During life, a second mutation occurs → accelerates growth further
      3. A third and fourth mutation occurs → formation of a tumor

      Types of mutations and their biological consequences

      A mutation is a permanent alteration in a parental DNA-sequence → a cell or organism. If it is in a parental organism, it is a hereditary mutation → is transferred to a child. If it occurs in a cell, the mutation is transmitted to the daughter cells.

      Classes of mutations:

      There are two classes of mutations:

      • Chromosome mutations: may affect the expression of many genes
        • Chromosome losses/gains
        • Translocations
        • Multi-locus deletions
      • Gene mutations: may affect the expression of a few genes
        • Deletions/insertions
        • Base pair substitutions
        • Frameshift mutations

      Chromosome mutations

      Multi-locus deletions:

      In case of multi-locus deletions, a fairly big part of a chromosome is deleted. This leads to a loss of function of the deleted alleles. Deletion on an autosomal chromosome leads to hemizygosity for multiple genes → only copy of the allele remains. Hemizygosity means that only 1 copy of the gene is left in 1 cell.

      Intragenic mutations:

      If there is a gene that consists of 5 exons, and there is a deletion of exon 2 and another deletion of exon 2 and 3, the mutation is intragenic. This doesn’t necessarily lead to loss of function, but most of the time, it does.

      This has mutagenic consequences:

      • If exon 2 is deleted, the reading frame doesn’t change → even though the protein may be smaller because it lacks a certain part, the total-protein will remain functional or at least partially functional
        • Exon 2 isn’t necessary for the function of the total protein
      • In case of exon 2+3 deletion, the reading frame does change → there is introduction of a stop codon → the protein becomes truncated and loses its function

      Gene mutations

      Base pair mutations over a gene can be distinguished in mutations which occur in:

      • Intron: most often have no consequences
      • Promoter: may affect transcription efficiency
        • Either increase or diminish
      • Splice site: may affect splicing
        • E.g. can cause exon skipping
      • Exon: may affect protein composition

      In a splicing consensus of intronic and exonic sequences the following is visible:

      • Intronic sequences always start with GT (GU in mRNA)
      • Intronic sequences always end with AG

      If this GT sequence is changed into an AT sequence, the exon isn’t recognized anymore → cannot encode its information into a protein. The same thing happens at the other end

      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC3: Cancer biology

      Mechanisms of Disease 2 HC3: Cancer biology

      HC3: Cancer biology

      Hallmarks

      There are 10 hallmarks of cancer which distinguish epithelial cells from carcinomas:

      1. Deregulating cellular energetics
      2. Sustaining proliferative signaling
      3. Evading growth suppressors
      4. Avoiding immune destruction
      5. Enabling replicative immortality
      6. Activating invasion and metastasis
      7. Inducing angiogenesis
      8. Resisting cell death
      9. Genome instability and mutation
      10. Tumor promoting inflammation

      Tumor promoting inflammation and genome instability and mutation are enabling characteristics → enhance the cancer development progress. The other hallmarks are fundamental changes in cell physiology.

      Sustaining proliferative signaling

      When proliferative signaling is sustained, growth is stimulated constantly:

      1. A growth factor binds to a growth factor receptor
      2. The growth factor receptor activates molecules, for instance Ras
      3. Active Ras phosphorylates kinases
      4. Kinases cause cell cycle progression
      5. Cell growth

      The phenotype is dominant → only 1 allele needs to be mutated. Some tumor cells:

      • Secrete their own growth factors
        • Become independent of growth factors from the outside
      • Modify their cell surface receptors
        • The receptors are constantly activated → don’t need growth factors anymore
      • Mutate their intracellular signal molecules
      • Mutate transcription factors
      • Mutate components of the cell cycle network

      Evading growth suppressors

      Normal body cells are in equilibrium with growth suppressors and growth promotors. There are several checkpoints for controlling this:

      • R-point
      • DNA-integrity checkpoint
      • Wnt signaling

      R-point:

      The R-point is the restriction point. Here, the Rb protein plays an important part → phosphorylation of Rb is necessary to release the restriction point. The Rb pathway is mutated in virtually all types of tumors. The Rb pathway can be inhibited by itself or by other means such as:

      • Loss-of-function mutations of growth inhibitors
        • TGF-b
        • INK4a
      • Gain-of-function mutations of growth factors
        • Cyclin D
        • CDK4

      Both mutations cause Rb to become hypophosphorylated.

      DNA-integrity checkpoints:

      DNA-integrity checkpoints monitor 3 things:

      • Whether the DNA is not too damaged
      • Whether there are not mutations
      • Whether the chromosome is properly attached to the spindle

      A key player in this is p53, which is also called the guardian of the genome. It can be activated by:

      • DNA-damage
      • Hyperproliferative signals
      • Telomere shortening
      • Hypoxia

      Activation of p53 leads to:

      • Cell cycle arrest
      • Senescence → not being able to replicate anymore
      • Apoptosis

      In more than 50% of all human tumors, p53 is mutated. p53 protective pathways are affected in more than 90% of all tumors. The following happens:

      1. Loss of p53
      2. Loss of DNA-integrity checkpoints → reduced apoptosis and senescence
      3. Proliferation of cells with DNA damage
      4. Mutations and chromosomal aberrations → genomic instability

      The Li Fraumeni syndrome is a hereditary mutation. It is a heterozygous mutation of p53 which leads to multiple primary tumors at young age. It is inherited dominantly. During development, in many tissues the second allele is lost → everyone gets cancer. It is recessive on cell level.

      Wnt signaling:

      In case of Wnt signaling, b-catenin is degraded by antigen presenting cells (APC):

      1. Wnt stimulation
      2. APC releases b-catenin
      3. b-catenin stabilizes and drives
      .....read more
      Access: 
      Public
      Mechanisms of disease 2 HC4: Cancer etiology

      Mechanisms of disease 2 HC4: Cancer etiology

      HC4: Cancer etiology

      Risk factors

      There are multiple risk factors for cancer. The most important ones are:

      • Age
        • The number of times a cell has divided
      • Genetics/hereditary: syndromes that lead to cancer, mainly in DNA repair
        • Lynch syndrome: predisposes to colon and endometrial cancer
        • Li Fraumeni: TP53 mutation
        • BRCA1 and BRCA2 → breast cancer
        • Cowden: PTEN mutation → breast, thyroid, uterine, renal and colon cancer
      • Environmental factors
        • Tobacco
        • Alcohol
        • Chronic inflammation
        • Diet
        • Hormones
        • Immunosuppression
        • Infection
        • Obesity
        • Radiation
        • Sunlight

      Risk factors can be split into controllable and non-controllable risk factors. Most are non-controllable, such as age and gender.

      Age

      Age is the greatest risk factor for cancer development. The peak of incidence of cancer is around 70 years. This isn’t equal for all cancer types → during puberty, the incidence of bone cancer rises due to fast growth.

      The more someone ages, the more times cells are divided and the more likely it is that mistakes are accumulated in DNA-strands. Because there are 3 billion base pairs, mistakes occur easily.

      Odds:

      Odds are generally against cancer:

      • There is a minority of mutations in oncogenes and TSGs
        • Mutations are much more likely to occur outside these genes
      • There are DNA repair mechanisms
        • Make sure that if mistakes happen during replication, they are repaired
      • Several features need to be acquired by cells to go from a healthy cell to a malignant cell
        • The 10 hallmarks of cancer

      Stem cells:

      Nevertheless, if someone lives long enough, many cell divisions occur and the chance of developing mutations becomes more likely. Because stem cells have to supply a constant amount of new cells throughout life, they divide a lot. This can cause mutations to accumulate at different sites.

      Hereditary cancers

      Hereditary means segregating in the family → transmission of cancer material. There is a familial high chance of genetic problems in the family, but the cause is unknown. Prevalence of hereditary cancers depends on the cancer type:

      • Hereditary: transmission of genetic material
        • The gene that causes it is known
      • Familial: most likely due to more than 1 genetic alteration
        • Vague clues that there is something in the family, but with what or what gene is unknown

      Environmental factors

      Environmental factors most often directly affect DNA molecules. Geographical and societal differences hint that environmental factors play a part in the development of cancer:

      • Prostate cancer is more common in developed countries → it is a disease of old age, and in developed countries people live longer
      • Liver cancer has something to do with hepatitis B infections → in developed countries hepatitis B vaccination is common → liver cancer is less prevalent
      • Cervical cancer is prevented in developed countries → cervical cancer is mainly caused by HPV

      Other examples of environmental factors are:

      • Alcohol → mouth, throat and esophagus cancer
      • Estrogen → breast and
      .....read more
      Access: 
      Public
      Mechanisms of disease 2 HC5: Hereditary aspects of cancer

      Mechanisms of disease 2 HC5: Hereditary aspects of cancer

      HC5: Hereditary aspects of cancer

      Risk of developing cancer

      The chance of getting cancer increases with age:

      • 8% by the age of 65
      • 25% by the age of 80
      • 32% by the age of 100

      If someone has a sibling with breast cancer and is 40-50 years old, the chance of developing breast cancer is 2x as high as usual. In case of colon cancer, the cancer is 3x as high. Among monozygotic twins, the risk is higher → genetic factors play an important role in the risk of developing cancer.

      Breast cancer

      Genetic factors:

      There are 14.000 new diagnoses of breast cancer per year in the Netherlands, of which 3.500 die every year. 13% have a first degree relative.

      Genetic factors play a role in whether or not someone develops breast cancer:

      • High/medium penetrant genes
      • Low penetrant genes/loci/SNPs

      There are 200 SNPs (single nucleotide polymorphisms) that increase the risk of breast cancer → per SNP, the increase is 0,1%, but multiple SNPs can be present at the same time.

      Genetic counseling:

      Genetic counseling is useful to:

      • Recognize patients and their families with inheritable cancer
      • Improve morbidity and mortality by early recognition and treatment
      • Psychosocial guidance and advice

      Cancer syndromes:

      Many cancer-causing genes have been discovered. Examples are:

      • RB gene
      • APC gene
      • Mismatch repair genes
      • BRCA1 and BRCA2
      • CDKN2A
      • MUTYH
      • BAP1

      Criteria for genetic testing:

      1 of the following situations has to be present to test for genetic breast cancer (BRC):

      • BRC occurs at <40 years
      • Bilateral BRC or multiple tumors in 1 breast with 1 tumor <50 years
      • First grade male with BRC
      • BRC <50 years and prostate cancer <60 years in the same branch of family
      • BRC <50 years and 1 or more first degree with BRC <50 years
      • BRC and 2 or more first and/or second degrees with BRC, of which at least 1 <50 years
      • Ovarian cancer irrespective of age
        • Have a 10% risk of a germline BRCA1 or BRCA2 mutation

      BRCA1 and BRCA2:

      BRCA1 and BRCA2 mutations are associated with inheritable breast cancer. They are DNA-repair genes. Chances of developing cancer if 1 of these mutated genes is present are high:

      • Breast cancer
        • BRCA1: 60-80%
        • BRCA2: 60-80%
      • Ovarian cancer
        • BRCA1: 30-60%
        • BRCA2: 5-20%

      BRCA mutation carriers undergo different types of surveillance, starting at early age:

      • Breast surveillance
        • 25-60 years: yearly MRI
        • 25-60 years: clinical breast examination
        • 30-60 years: yearly mammography
        • 60-75 years: population screening
      • Ovaries
        • 35-40 years: prophylactic adnex extirpation
          • Ovaries and fallopian tubes are removed

      For males with a BRCA2 gen, the risk of developing prostate cancer is 2-4x as high.

      They undergo a blood test for PSA at the GP, every 2 years starting at the age of 45.

      Li Fraumeni syndrome:

      In case of Li Fraumeni syndrome (LFS), there is a mutation of the TP53 gen. LFS is characterized

      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC6: Cancer and genome integrity

      Mechanisms of Disease 2 HC6: Cancer and genome integrity

      HC6: Cancer and genome integrity

      Genome instability and mutation

      Many cell divisions are required in embryonic development and during human life. Daughter cells inherit identical DNA. Cell division requires accurate DNA duplication and takes approximately 5 hours. 6 x 109base pairs are copied almost flawlessly → there is less than 1 error per division.

      Mutation repair:

      If not repaired, DNA lesions can lead to mutations. Mutations can arise during regular DNA replication → DNA replication errors. Base mis-incorporation can occur, for example, a T can be inserted opposite of a G. Originally, there are 60.000 errors per cell division. However, there are several processes to correct such mistakes:

      • Proofreading by DNA polymerase + exonuclease
        • Exonuclease activity makes it possible for DNA polymerase to misincorporate a base, go back to the former situation and try it again
        • Germline mutations in POLE and POLD1 (the main polymerases that replicate DNA) predispose to colorectal adenomas and carcinomas
        • Reduces misincorporation by DNA polymerase from 10-5to 10-7→ 600 errors per cell division
      • Mismatch repair
        • Removes the newly synthesized strand, so that polymerase can try it again
          • Occurs if exonuclease has not been able to remove the incorrect base
        • Reduces the error rate during DNA replication from 10-7 to 10-9 or 10-10 → there is less than 1 error per cell division on undamaged DNA

      Lynch syndrome:

      Lynch syndrome is a hereditary colon cancer. In case of Lynch syndrome, there is a mismatch repair deficiency. Several mutations predispose to developing Lynch syndrome, such as mutations in:

      • MLH1
      • MSH2
      • MSH6
      • PMS2

      Replication of damaged DNA

      Threats to genome stability:

      Humans are continuously exposed to sources which damage DNA. DNA damaging agents can form threats to genome stability:

      • Lifestyle
      • Environmental or industrial
      • Medical application
      • Food source

      Sunbathing:

      In case of sunbathing there is exposure to UV-light. This causes replication blocks → a base is linked to another base → DNA polymerase stops because it cannot recognize a base. The DNA polymerase stops replicating → causes cell death.

      A number of polymerases can bypass DNA damages → translesion synthesis polymerases (TLS). Because TLS can replicate damaged DNA, cell death is prevented. Sometimes these polymerases make errors, leading to a next round of replication for mutation.

      POLη is the translesion polymerase bypassing mutation due to UV-damage. This is one of the most precise translesion polymerases → has evolved a lot. If another translesion polymerase bypasses the DNA damage, the chance of errors is higher.

      A germline homozygous mutation of POLη leads to xeroderma pigmentosum variant-patients. Because other translesion polymerases have to do the work, these patients have sun-damaged skin and can easily develop skin cancer.

      Endogenous sources:

      Human cells also produce DNA damage every day:

      • Production of free radicals
      • Chemical instability
        • 10.000 bases per cell per day are lost
        • Depurination
          • Especially purines are prone to hydrolyse
          • The base is lost → suddenly there is no information during replication
        • Deamination: cytosine is methylated
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC7: Clinical relevance of genetic repair mechanisms

      Mechanisms of Disease 2 HC7: Clinical relevance of genetic repair mechanisms

      HC7: Clinical relevance of genetic repair mechanisms

      Introduction

      BRCA and Lynch syndrome occur very frequently in the population. Both syndromes are caused by DNA repair genes.

      Even though germline mutations are equally distributed through the genome, hereditary cancer cases associate with syndromes caused by mutations in DNA repair genes. On the other hand, cancers related to inherited APC-mutations are also relatively frequent.

      Damage and clinical prognosis

      There is an association between DNA repair defects and clinical prognosis. Lung cancers have much more mutations than breast cancers. However, if there are more mutations, a tumor doesn’t necessarily have a worse prognosis. There are several reasons for this:

      • There can be too much DNA damage in cancer cells → essential cancer genes are hit → the cell dies due to excessive damage
      • Point-mutations are not the only alterations that occur in a cancer cell → tumors with very few mutations may have very abundant chromosomal mutations

      Colorectal cancer genetics and prognosis:

      Colorectal cancer can develop in several different ways:

      • A minority develops under very specific defects in the DNA repair machine
      • A small proportion (less than 5%) develops under deficiencies in the proofreading domain of polymerase chains → accumulate to a lot of mutations in their coding gene
      • 15-20% develops under the context of mismatch repair deficiency
      • A large majority has a low number of mutations in their genome but a large chromosomal instability

      Even though MMR-deficient (mismatch repair deficient) or POLE-mutated (polymerase mutated) tumors have more mutations than MMR-proficient tumors, they have a better prognosis up to stage 3. In stage 4, the prognosis is equally bad for all tumors. This means that more mutations actually result in a better prognosis. This can be explained by:

      • There being too much damage → affects the tumor cells
      • Chromosomal instability leading to a worse prognosis than mutations

      MMR-deficient and POLE-mutated tumors have a lot of lymphatic infiltrates in their tumors.

      Endometrial cancer and prognosis:

      In endometrial cancer, MMR-deficient and POLE-mutated tumors are also present. Here, POLE-mutated patients do better than patients who are MSS (microsatellite stable) or MSI (microsatellite instable). MSI-patients are MMR-deficient and MSS-patients are MMR-proficient. POLE-mutated and MSI patients have more lymphocyte infiltrates, which mainly consist of CD8 T-cells, than MSS-patients.

      BRCA status and breast cancer prognosis:

      Approximately 10% of all breast cancers carry defects in the BRCA genes, of which half are due to germline mutations. There has been a long debate about whether there is a relation between prognosis in breast cancer and the status of BRCA mutations. Recently, a big study has shown that there is no difference between BRCA-positive and BRCA-negative patients.

      Arguments supporting an association between BRCA1 mutations and worse prognosis are:

      • 70-90% of BRCA1 related tumors are triple-negative
        • Don’t express estrogen-receptors or HER2-receptors

      Arguments against an association between BRCA1 mutations and a worse prognosis are:

      • BRCA-related cancers are detected at earlier stages
      • If looking at triple-negative tumors, BRCA mutations are associated with
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC8: General principles: diagnostic pathology

      Mechanisms of Disease 2 HC8: General principles: diagnostic pathology

      HC8: General principles: diagnostic pathology

      Biopsies

      A spiculated mass is a mass with spikes going into the surrounding tissue. Benign lesions generally don’t grow in this type of fashion → a biopsy needs to be taken.

      Fine needle aspiration cytology:

      FNAC stands for fine needle aspiration cytology:

      1. A little bit of tumor is taken from the lesion with a needle
      2. The tumor is spread out on a small glass
      3. Color is added → rapid stain-RAL
        • This is a stain which takes 40 seconds

      This isn’t a histology biopsy, because the cells are loose.

      Visible cells:

      Biopsies can be taken to test for breast cancer. Various cells become visible:

      • Tumors
      • Cells without a nucleus → erythrocytes
      • Irregular cells vary in size

      In a normal situation, the following is visible:

      • Low cellularity
      • Cohesive cell-groups
        • Cells are small and have the same size and shape
      • Small cells
        • Maximal 1,5x the size of an erythrocyte

      Signs of cancer cells are:

      • Large, variably shaped nuclei
        • Hyperchromasia
          • The nucleus is darker
        • Polychromasia
      • Many dividing cells → disorganized arrangement
        • Cells come loose from epithelia
      • Variation in size and shape
        • Polymorphism
        • Pleomorphism
      • Loss of normal features

      Malignancy

      Several important terms are:

      • Benign: not harmful
      • Malignant: harmful
      • Atypia: a clonal proliferation
      • Ductal carcinoma in situ (DCIS): not yet cancer
        • The cells look malignant, but this doesn’t actually prove that the lesion is malignant

      A tumor is malignant when:

      • Invasion occurs → malignant tumors recognize no anatomic boundaries
      • Cells have the ability to metastasize → can invade blood and lymphatic vessels

      Breast carcinoma:

      A carcinoma can be invasive or in situ. Which of the 2 is the case cannot be seen on a biopsy:

      1. Normal breast tissue is made of fat and duct
        1. Ducts transport milk to the nipple
        2. A duct contains an epithelial luminal layer which can transport and form the milk
      2. When hyperplasia occurs in those ducts, the cell layer is proliferating within the duct
      3. Atypia can occur in the proliferating cell population, which can lead to DCIS (ductal carcinoma in situ)
      4. DCIS: the carcinoma is still in the duct and hasn’t travelled to a different location
        • DCIS cannot metastasize → isn’t a deadly disease
      5. When the carcinoma travels somewhere else, it becomes an invasive ductal
        • Invasive ductals can metastasize

      DCIS and invasive ductals cannot be distinguished on a microscopy slide → malignancy cannot be distinguished from carcinoma in situ. A core biopsy is necessary to do this. It also can be useful to palpate the lymph nodes in the axilla. Tumor cells in lymph nodes proof that the disease is malignant. An adenocarcinoma is a carcinoma in glandular tissue. It can metastasize in lymph nodes.

      Macroscopical aspects:

      Macroscopical aspects of malignancies are:

      • Different from the surrounding normal tissue
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC9: Nomenclature and grading of cancer

      Mechanisms of Disease 2 HC9: Nomenclature and grading of cancer

      HC9: Nomenclature and grading of cancer

      Nomenclature

      Mesenchymal tumors:

      There are many kinds of mesenchymal tumors:

      • Fat: lipo-
      • Bone: osteo-
      • Fibrous: fibr-
      • Cartilage: chondro-
      • Blood vessels: hemangio-
      • Muscle: leiomyo-/rhabdomyo-

      These tumors can be benign of malignant:

      • Benign: -oma
        • Cells are nicely organized
      • Malignant: -sarcoma
        • Sarcoma means flesh → malignant tumors are fleshy
        • Cells aren’t organized

      Epithelial tumors:

      Epithelium is the coverage and lining of the body. Tumors can be:

      • Glandular or ductal
      • Squamous and stratified

      Epithelial tumors can be benign of malignant:

      • Benign
        • Adenoma
          • Cystadenoma: a cyst originating from a duct
        • Papilloma
      • Malignant
        • Carcinoma
          • Adenocarcinoma: carcinoma in the colon
          • Papillary carcinoma

      Melanocytes:

      Melanocytes can be benign or malignant:

      • Benign
        • Nevus: a mole
      • Malignant
        • Melanoma
          • In this case, “-oma” stands for a malignant disease

      Testicular epithelium:

      Tumors in testicular epithelium are always malignant → seminomas.

      Totipotent cells can form all kinds of tissues. They are also located in the testis and ovaries. Tumors are called teratomas:

      • Benign
        • Mature teratoma
      • Malignant
        • Immature teratoma/terato-carcinoma

      In women, teratomas are often benign. They can contain all types of tissues, even complete teeth.

      Salivary glands:

      Salivary glands have many different features. Tumors in salivary glands can vary greatly. They can be benign or malignant:

      • Benign
        • Pleiomorphic adenoma
          • Has a high recurrence
        • Many others
      • Malignant
        • Mucoepidermoid carcinoma
        • (Basal) adenocarcinoma
        • Acinic cell carcinoma

      Grading

      Grading defines the impact of grade and stage in different malignancies and can explain the differences between these features. A grade can be used as a biomarker, which is:

      • Prognostic: for example of the chances of metastasis
      • Predictive: for example of the chances of response to certain types of therapy

      For all tumor types, there are different grading systems.

      Grading of benign tumors:

      There are 4 grades for benign tumors:

      • Grade 1: well differentiated → low grade
      • Grade 2: moderately differentiated → intermediate grade
      • Grade 3: poorly differentiated → high grade
      • Grade 4: undifferentiated → high grade

      Bloom and Richardson:

      Bloom and Richardson made grades which can be used to predict the survival rate, for instance for breast cancer:

      • Grade 1 → 75% after 5 years
      • Grade 2 → 53% after 5 years
      • Grade 3 → 31% after 5 years

      Fuhrman grading system:

      The Fuhrman grading system can be used in renal cell carcinomas. These cells have clear cytoplasms. The bigger the nucleoli (dots in the nuclei), the higher the grade. There are 4 grades in total.

      Gleason scoring system:

      The Gleason scoring system can be used in case of prostate cancer. It is based on the amount of nicely formed ducts:

      • Grade 1: nearly normal cells
      • Grade 2: some abnormal cells, loosely packed
      • Grade 3: many abnormal cells
      • Grade
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC10: General principles: metastasis

      Mechanisms of Disease 2 HC10: General principles: metastasis

      HC10: General principles: metastasis

      Historical perspective

      Metastasis is Greek word, which in medical use describes the shift of disease from one part of the body to another. It is mainly used in context of cancer.

      Jean Claude Recamier:

      In the 19th century, Jean Claude Recamier recognized that cancer can spread from a primary tumor.

      Stephan Paget:

      Stephan Paget came up with the seed-and-soil theory. He stated that the growth of a tumor is not a matter of chance, but that there must be an affinity between the tumor cells and the site where they are metastasizing.

      Sister Mary Joseph node:

      Sister Mary Joseph noticed that a tumor in the umbilicus usually is a metastasis of a tumor elsewhere → a sister Mary Joseph node is a secondary lesion in 1-3% of abdominal cancers. Its metastatic route is unclear.

      James Ewing:

      In 1929, James Ewing states that metastatic dissemination occurs by purely mechanical factors.

      Mechanistic theory:

      The mechanistic theory states that mechanical forces and circulatory patterns between the primary tumor and the secondary site account for organ specificity. This forms a contradiction to the seed-and-soil theory. In the end, both theories proved to be correct → depends on the cancer type.

      Bernard Fisher:

      In 1965, Bernard Fisher described breast cancer as a systemic disease. He stated that early-stage breast cancer can be more effectively treated by lumpectomy, in combination with radiation therapy, chemotherapy and/or hormonal therapy than by radical mastectomy. Mastectomy is an extremely invasive procedure where the breasts, muscles and lymph nodes are removed. If metastasis has already occurred, such treatment isn’t useful.

      Isiah Fidler:

      In 1975, Isiah Fidler stated that metastasis is the result from the survival of only a few tumor cells. The process of metastasis and invasion is comprised of several stages:

      1. A primary tumor is proliferating and generating clonal heterogeneity
      2. Angiogenesis takes place
      3. Tumor cells start migrating from the primary site
      4. Tumor cells enter lymphatic and blood vessels → are transported throughout the body
      5. Tumor cells start interacting with other cells, including immune cells
      6. When they arrive at different organs, tumor cells leave blood vessels
      7. Tumor cells grow in a new environment

      This is a complex process which consists of different steps. A tumor clone needs to acquire all these features, which is quite difficult. One of the ways they can do this is through genomic instability. Once a tumor cell has acquired the 10 hallmarks of cancer, it can metastasize.

      Robert Weinberg and Jean Paul Thiery:

      In 2002, Robert Weinberg and Jean Paul Thiery introduced the concept of EMT (epithelial to mesenchymal transition). This concept is specifically applied to epithelial tumors:

      1. Epithelium is located on top of the basal membrane, mesenchymal cells are located beneath the basal membrane
      2. If epithelial tumors want to grow in the mesenchymal section, they need to acquire features that are similar to mesenchymal cells to be able to survive the environment

      Mechanisms

      EMT:

      EMT most often refers to functional characteristics gained during malignant progression rather than

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC11: General principles: molecular diagnostics

      Mechanisms of Disease 2 HC11: General principles: molecular diagnostics

      HC11: General principles: molecular diagnostics

      Molecular diagnostics

      Molecular diagnostics is a subspecialty of pathology that utilizes molecular biology techniques to:

      • Detect normal and disease states
      • Make a prognosis
      • Identify druggable targets

      Molecular biology techniques utilize DNA, RNA and enzymes that interact with nucleic acids to understand biology at a molecular level:

      • KRAS mutations
      • EGFR deletion
      • EML4-ALK or ROS translocations in NSCLC
      • Et cetera

      Therapeutic targets

      (Proto)oncogenes have several therapeutic targets based on the hallmarks of cancer:

      • Growth factor
      • Growth factor receptor
      • Intracellular signal transduction pathway
      • Cell cycle regulators
      • Transcription factors
      • Anti-apoptotic factors

      Abnormal activation of these targets leads to cancer. This activation can be caused by:

      • Dominant mutations at cellular level
      • Single mutations → 1 allele is sufficient

      Aneuploidy:

      In a tumor cell, chromosomes become heavily dysregulated. This can even lead to entire chromosomes changing. When the chromosome number changes, it is called aneuploidy.

      Type of mutations

      Activating mutations can be divided into 3 categories:

      • Translocation
      • Point mutations
        • Missense mutations
          • An amino-acid is changed into another amino-acid
        • Nonsense mutations
          • A stop codon is introduced instead of an amino acid → truncated protein
        • Silent mutations
          • Even though a base pair is changed, it still codes for the same amino acid
        • Indel/frameshift mutations
          • A base is deleted and the next nucleotide is taken for translation
      • Amplification
        • A normal gene becomes amplified and has multiple copies

      Use:

      Molecular diagnostics are useful to diagnose several things:

      • Tumor phenotype
      • Malignancy
      • Origin of the tumor
      • Heredity

      Therapy:

      Therapy can consist of:

      • Personalized medicine
      • Treatment of cancer
      • Drugs targeting specific mutations/pathways

      Material available for testing:

      The vast majority of tissue is frozen and put into paraffine slides. This can be used for histological images → the location of tumor cells can be identified.  

      Tumor heterogeneity

      A tumor consists of:

      • Parenchym
        • Real cancer cells/neoplastic cells
          • Have the actual mutations
          • Surrounded by macrophages, fibroblasts, etc.
      • Stromal cells
        • Connective tissue and vessels
        • Different type of normal cells

      Lots of lymphocytes are visible around tumor cells.

      Detection of mutations

      A tumor is a mix of neoplastic cells and supporting “normal” cells. The tumor cell percentage is the estimated % of tumor cells. Tumor cells are genetically instable and have a large variety. They can be distinguished as follows:

      • Normal cell
        • 2 wild type alleles
      • Tumor cell
        • 1 wild type allele
        • 1 mutation

      Testing

      There are 87 FDA approved drugs. For each drug, there is a molecular target which needs to be tested in the lab for 45 different disease indications. Every drug acts upon 1 hallmark of cancer.

      BRAF:

      A BRAF melanoma is caused by a BRAF mutation of the BRAF inhibitor. The following is found:

      • Biopsy: neoplastic tissue with <1% tumor cells
      • Microdissection: approximately 10% neoplastic
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC12: How did cancer become the emperor of all maladies?

      Mechanisms of Disease 2 HC12: How did cancer become the emperor of all maladies?

      HC12: How did cancer become the emperor of all maladies?

      Overview

      Cancer is the emperor of all maladies, the king of all terrors. The attention to and visibility of cancer is growing:

      1. In Greek and Roman times, cancer was visible but not so prominent
      2. More and more research of tissues is done
      3. Around 1900, cancer became a new interest → a public health concern
      4. The interest in occupational and environmental causes and complexity started growing

      Cancer has become a more and more visible disease over the last two centuries. It has developed into the disease of modern times. The disease became a major object of medical, social, economic and political concern.

      17th century

      In the 17th century, breast cancer was seen as a female disease because only in women it was visible on the outside. Breast amputation was preformed, which had to be done very fast. Of course, men also had breast cancer, but they attributed the signs to something else than a tumor in the body. Breast cancer was connected with lust in sexuality because no breast cancer was found in nuns.

      19th century

      In the 19th century, a first turning point in cancer history occurred. It was stated that diseases weren’t located throughout the entire body, but could emerge from certain tissues and other parts. There was a localist approach focused on cells, tissues and organs. Doctors opened up a body and found tumors inside. Rudolf Virchow made an important statement: “all cells arise from other cells and malignant cells grow too fast”.

      20th century

      Early 20th century:

      In 1900, the words cancer genetics and oncologist did not exist. Chemotherapy wasn’t present, but a cancer hospital was. Radiation therapy could already be preformed. In 1913, the Netherlands Cancer Institute (NKI) was established.

      In the early 20th century, cancer increasingly became a public health problem. This formed a second turning point. It statistically came more to the surface because the life expectancy started to rise. People started thinking more about prevention and control and saw it as “an enemy we have to fight”. Several patterns became visible, such as apprentice chimney sweepers getting scrotal cancer. Between the 1930s and 1960s, surgery underwent huge developments.

      Late 20th century:

      After World War II, the complexity, risk and genetics of cancer became more and more visible. The erosion of cancer as a social taboo started.

      Between the 1960s and 1980s, the relation between environment, behavior and cancer became increasingly visible. This formed a third turning point. For instance, smoking used to be very normal but caused many cases of cancers. Open minded doctors where needed, because the industry would put profit before health. They would create doubt about the relationship between smoking and lung cancer → “more doctors smoke Camels than any other cigarette”. Cancer was described as a single monolithic entity, which isn’t true.

      Access: 
      JoHo members
      Mechanisms of Disease 2 HC13: Heterogeneity in cancer

      Mechanisms of Disease 2 HC13: Heterogeneity in cancer

      HC13: Heterogeneity in cancer

      Types of heterogeneity

      In cancer, there are 3 types of heterogeneity:

      • Inter-patient: between patients
      • Inter-tumor: between tumors
      • Intra-tumor: within the tumor

      Examples are:

      • Well/moderately differentiated versus poorly differentiated
      • Tumors with a strong T-cell infiltration versus with a poor T-cell infiltration
      • Cancer cells are different when they are at the core than when they are at the invasive margin
        • Because the environment is different
          • For example the amount of immune cells or the access to oxygen
      • The more clones there are in a tumor, the more likely the chance that 1 is able to metastasize

      Heterogeneity forms a major issue for the treatment of cancer.

      Intra-tumor and inter-tumor heterogeneity

      Intra-tumor heterogeneity:

      There are 2 origins of intra-tumor heterogeneity:

      • Every time a tumor cell divides, it is an opportunity for a new clone to arise
      • DNA repair deficient tumors are likely to be more clonal heterogenous

      The fact that a new clone evolves, doesn’t mean it will survive:

      • It has to compete with other clones
        • Competition for nutrients/oxygen
        • Proliferation
      • It has to evade the immune system

      Inter-tumor heterogeneity:

      Inter-tumor heterogeneity is a morphological heterogeneity between tumors → one tumor is well/moderately differentiated while the other is poorly differentiated.

      Heterogeneity becomes clear when the protein expression is observed. For instance, 1 tumor can have high levels of PD-L1 expression while the other doesn’t. Tumor cells which are negative for PD-L1 respond negative to the treatment.

      Not only the tumor cells present in tumors can differ, but also other cells, for instance immune cells.

      Origins of cancer cell heterogeneity:

      Heterogenic tumors arise as follows:

      1. Epigenetic changes occur during cell division
      2. A clone that is able to evade the immune system survives and keeps dividing
      3. The clone keeps getting better at evading the immune system

      Of all the clones, 1 clone can remain resistant to therapy.

      Clonal cooperation:

      Clonal cooperation refers to characteristics of cancer provided by distinct cancer cell clones. Different clones, which all are present in the same tumor, have different functions:

      • Clone A: tumor proliferation
      • Clone B: angiogenesis
      • Clone C: invasion

      Clonal competition:

      Aside clonal cooperation, there is competition between cancer cells. Cells compete for nutrients, space and oxygen. Many clones that are generated during tumor progression do not survive. This competition is driven by selective pressure, which is driven by either external factors or intrinsic features of cancer cells:

      • Immune recognition
      • Competition of nutrients/oxygen
      • Inter-clonal competition

      Inter-patient heterogeneity

      Different genes are mutated in different cancers. Only TP53 mutations occur in nearly every type of cancer. The large majority of mutated genes differs considerably from one tumor to the other tumor type. They play different roles in healthy tissues. This inter-patient heterogeneity has consequences for the treatment.

      An example is HER2, which is mainly expressed in breast and urothelial cancer. These cancer types can be treated with trastuzumab.

      Different mutation profiles:

      Inter-patient heterogeneity can

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC14: Cancer immunity and immunotherapy

      Mechanisms of Disease 2 HC14: Cancer immunity and immunotherapy

      HC14: Cancer immunity and immunotherapy

      Immune responses in cancer

      The incidence peak of cancer is around the 6th decade of life. Therefore, it isn’t important in the evolution or in shaping the immune system → mainly occurs after reproductive age.

      In the beginning of the 20th century, a number of scientists proposed a theory of immune surveillance, which later was further confirmed:

      1. If a mouse is injected with a carcinogen, a tumor starts developing
      2. If the tumor is injected into another mouse, the tumor continues developing
      3. If the tumor is first subjected to radiation and vaccinated into a mouse, the tumor stops growing

      This shows that the immune system plays an important role in tumor growth.

      Tumor-infiltrating immune cells

      The presence of certain immune cells correlates with both improved and worse tumor behavior and prognosis.

      T-cells:

      T-cells are associated with both a good and bad prognosis, depending on the type:

      • Presence of CD8 T-cells correlates with a good prognosis
      • Presence of tertiary lymphoid structures are a good sign
      • Presence of regulatory T-cells are associated with a worse prognosis → immune suppressive role

      Macrophages:

      Macrophages are often associated with immunosuppressive activity. They can produce cytokines like IL-10 or TGF-β, which counteract immune activity. There are 2 types of macrophages:

      • M1: pro-inflammatory → better prognosis
      • M2: immunosuppressive → worse prognosis

      The majority of macrophages in tumors are M2 → make a bad prognosis.

      Genetic instability and immune recognition

      Immune recognition:

      When a cell is targeted by carcinogens such as UV-radiation or cytotoxic drugs, the following happens:

      1. Damage response pathways are activated, such as:
        • ATM
        • P53
      2. When these proteins are activated, they induce the expression of a number of stress molecules which go to the surface of cells
      3. The proteins are recognized by NK-cells or CD8 T-cells
      4. The target tumor cell is eliminated

      However, a lot of these pathways are shut down in tumors. This causes the immune system to not be able to recognize tumor cells through these mechanisms anymore.

      Antigen processing and presentation:

      HLA-II molecules present proteins which are cut into peptides to cells. Although the main function of B-cells is antibody production, they can also produce HLA-I and HLA-II. Antigen presenting cells are:

      • B-cells
      • Dendritic cells
        • Professional antigen presenting cells
      • Macrophages

      These cells have HLA molecules:

      • HLA-I: presents to CD8 → kills the target cell
      • HLA-II: presents to CD4 → activates B-cells to kill the pathogen

      Both dendritic cells and macrophages have phagocytic capacity. Dendritic cells are the most effective APCs → provide the right co-stimulatory signal.

      However, not all peptides/proteins are presented to T-cells. This depends on the HLA alleles present in the immune system, which differs per person. Furthermore, even if a peptide is presented, not all peptides can be recognized by T-cells → an individual must have a compatible T-cell receptor that can recognize a specific HLA/peptide complex.

      Antigens

      Antigens presented to immune cells by tumor cells are:

      • Self-proteins:
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC15: Framework oncology and staging

      Mechanisms of Disease 2 HC15: Framework oncology and staging

      HC15: Framework oncology and staging

      Tumor grading

      A tumor grade reflects intrinsic biological behavior of tumors. In general, a low grade is less aggressive. It is necessary to grade a tumor for treatment and prognosis:

      • Lower grade → better prognosis
      • Higher grade → worse prognosis

      Tumors are graded based on the microscopic appearance of cancer cells. Dependent on the tumor, there are 2-4 degrees of severity. An example is:

      • GX: grade cannot be assessed
        • Undetermined grade
      • G1: Well-differentiated
        • Low grade
      • G2: moderately differentiated
        • Intermediate grade
      • G3: poorly differentiated
        • High grade
      • G4: undifferentiated
        • High grade

      Breast cancer grades:

      The amount of grades differs per tumor type. In case of breast cancer, there are only 3 grades. These grades can be linked to the expected 5 year survival rates:

      • Total: 91-95%
      • Grade I: 75%
      • Grade II: 53%
      • Grade III: 31%

      Staging systems

      Tumor staging is used to determine the extent of disease spread in patients:

      • Solid tumors
        • Local spread
        • Through lymphatics (lymphogenic)
        • Distant (hematogenic)
      • Haemotological malignancies
        • Tells something about the disease extent and influence on normal bone marrow function

      Cancer staging is useful for:

      • Planning treatment
      • Estimating prognosis
      • Identifying clinical trials/studies
      • Making a comparison between institutes
      • Communication
        • Staging is a universal language

      There is no unique staging system. However, most staging systems do consist of several common elements:

      • Location of the primary tumor
      • Tumor size and number of tumors
      • Lymph node involvement
        • Spread of cancer into the lymph nodes
      • Cell type and tumor grade
        • How closely the cancer cells resemble normal tissue
      • Presence or absence of distant metastasis

      In general, the higher the stage, the worse the prognosis.

      TNM staging principles:

      The TNM staging principles is the most used staging system for solid tumors:

      • T-status: extent of the tumor
        • T1-T4
          • The larger the tumor, the higher the T
        • Size of the tumor and locally organ involvement
      • N-status: regional lymph node involvement → absence or presence of metastasis
        • N0: no lymph nodes are involved
        • N1: 1 or 2 lymph nodes are involved
        • N2: infra- or subclavicular lymph nodes are involved
      • M-status: distant metastasis
        • M0: absent
        • M1: present

      The rules per tumor differ → prefix modifiers can be useful:

      • cTNM → c = clinical
        • Before treatment
        • Mostly based on imaging or physical examination
      • pTNM → p = histopathological
        • After surgery
      • ypTNM → yp = pathological after pretreatment
        • After chemotherapy, but before surgery
        • There is a large difference between the c and p stage
      • TNMx → x = not classified
        • Nothing is known about the tumor

      For instance, a PA report states the following:

      • Ductal adenocarcinoma
      • Grade 3
      • Diameter: 1,2 cm
      • Free excision
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC16+17: Pharmacology I&II

      Mechanisms of Disease 2 HC16+17: Pharmacology I&II

      HC16+17: Pharmacology I&II

      WHO 6STEPS

      To determine what therapy is effective, it is necessary to make a therapeutic plan. This can be done according to the WHO 6STEPS method:

      1. Step 1: evaluate the problems of the patient
      2. Step 2: identify the goals of therapy
      3. Step 3: list the treatment options (indication related)
      4. Step 4: provide the rationale for the best treatment for this patient
      5. Step 5: write a definitive therapy plan (prescription)
      6. Step 6: determine the monitoring parameters/follow-up

      To make a therapeutic plan for a specific patient, it is necessary to understand:

      • The pathology of the disease
      • The target sites of therapy
      • The mechanisms of actions of a drug

      Determining tumor treatment:

      Cancer is a heterogenous disease → it has different causes and requires different treatments. There are 3 main forms of cancer treatment:

      • Surgery
      • Radiotherapy
      • Pharmacological therapy

      To make a patient-specific therapeutic plan, the following needs to be determined:

      • Type of tumor
      • Growth velocity
      • Metastases

      Sometimes, curation isn’t possible anymore and treatment is palliative. Palliative treatment consists of:

      • Pain management
      • Supportive care

      Core medication list

      Groups of medicines:

      There are 4 groups of pharmacological therapy which can be used as cancer treatment:

      • Cytostatics
      • Hormones
      • Immunomodulators
      • Target therapy

      Core medication list:

      The core medication list consists of the most used drugs for cancer. It is necessary to know the:

      • Indication
      • Mechanism of action
      • Relevant side effects
      • One or 2 drug examples

      The core medication list is built up as follows:

      • Cytostatic drugs
        • Alkylating and related agents
          • Cyclophosphamide
          • Cisplatin
        • Antimetabolites
          • Methotrexate
          • Fluorouracil (5-FU)
          • Cytarabine
        • Antimitotic
          • Paclitaxel
        • Topoisomerase-inhibitors
          • Doxorubicin
      • Hormonal drugs
        • Antihormones
          • Tamoxifen
        • Aromatase-inhibitors
          • Anastrozol
        • Progestagens
          • Megestrolacetate
        • LHRH-analogues
          • Gosereline
      • Target therapy
        • Inhibitors of EGF-2 signaling
          • Trastuzumab
          • Cetuximab
        • Inhibitors of VEGF-signaling
          • Bevacizumab
        • Inhibitors of CD20/CD50 signaling
          • Rituximab
        • Tyrosine kinase inhibitors
          • Imatinib
        • (Immune) checkpoint inhibitor
          • Nivolumab
      • (Anti-)coagulants/immunomodulators
        • Thrombocyte aggregation inhibitors
          • Clopidogrel
          • Acetylsalicylic acid (aspirin)
        • Vitamin K antagonist
          • Acenocoumarol
        • Heparin
        • Antifibrinolyticum
          • Tranexamicacid
        • Thrombolyticum
          • Streptokinase
        • Coagulation factor
          • Thrombin
        • Xa inhibitor
          • Dabigatran
          • Apixaban

      Cytostatics

      Cell division:

      In normal cells, cell division is regulated by growth stimulating factors and growth inhibiting factors:

      • Growth stimulating factors → transcription of proteins that stimulate cell division
      • Growth inhibiting factors → transcription of proteins that inhibit cell division

      In cancer cells, growth is out of control. This is caused by:

      • Upregulated transcription of proteins that stimulate cell division
      • Downregulated transcription of proteins that inhibit cell division

      The cell cycle consists of 5 stages:

      1. G0
      2. G1: growth and preparation of DNA synthesis
      3. S: DNA replication
      4. G2: growth and preparation of mitosis
      5. M:
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC18: Biomarkers for early detection of cancer

      Mechanisms of Disease 2 HC18: Biomarkers for early detection of cancer

      HC18: Biomarkers for early detection of cancer

      Biomarkers

      Cancer can be diagnosed based on:

      • Physical examination
      • Blood tests
      • CT-scans
      • Biopsies to obtain tumor tissue

      A biomarker refers to a measurable indicator of some biological state or condition. Humans shed particles into the bloodstream or environment as evidence of their presence in a particular location:

      • Small parts of cells
        • Microparticles
        • Exosomes
      • Proteins
      • Unique chemicals
      • DNA and RNA

      Biomarkers are measured and evaluated to examine:

      • Normal biological processes
      • Pathogenic processes
      • Pharmacologic responses to a treatment
        • Therapeutic intervention

      An ideal tumor marker for diagnosis:

      • Should have great sensitivity, specificity and accuracy in reflecting the total disease burden
      • Should be prognostic of the outcome
      • Should be predictive of tumor recurrence
      • Should be predictive of effectiveness of anti-cancer treatments

      Samples for biomarker detection are:

      • Blood, urine or other body fluids
      • Tissues

      Tumor cells and products circulate in blood and are easily detectable. However, tissue forms the issue. There is a limited access to tissue:

      • Archival tissue is not obtained at the time of clinical decision
      • Accessible metastatic tissue may not be representative

      Circulating tumor cells

      Because tissue access is limited, there is an increased interest in circulating tumor cells (CTCs). CTCs shed by primary and metastatic tumors can be used as “liquid biopsies”, providing real-time information about the patient’s current disease state. Molecular profiling in CTCs can be used for patient selection to stratify for targeted therapy or serve as well-defined treatment targets.

      Clinical applications

      Potential clinical applications of CTCs are:

      • Blood testing using CTCs may aid in cancer diagnosis
      • May serve as prognostic and predictive biomarkers
        • Changes in CTC counts could indicate sensitivity or resistance to anti-cancer therapy monitoring
      • CTC enumeration may estimate tumor burden and tumor invasiveness
      • Investigational platform to eludicate tumor biology
        • Whole genome/transcriptome sequencing

      Microparticles:

      CTCs are not whole tumor cells → they are circulating biomarkers. CTCs are very rare in the blood of cancer patients, ranging from 1 to over 1000 in 10 ml blood samples, which usually contain 50-100 billion red and white blood cells.

      Microparticles from a tumor are released into the bloodstream. Microparticles are:

      • CTCs
      • Genomic DNA
      • miRNA
      • Microvesicles (MV)
      • Microparticles (MP)

      Clinical uses

      Clinical uses of biomarkers are:

      • Screening
      • Making a diagnosis
      • Marker of prognosis
      • Monitoring of treatment efficacy
      • Detection of recurrence of the disease

      Screening:

      A biomarker for screening must be:

      • Highly specific (for a single type of cancer) → minimize false positive and negative
      • Able to clearly reflect the early stage of disease
      • Easily detectable without complicated medical procedures
      • Cost effective

      Monitoring of treatment efficacy:

      A biomarker for monitoring treatment efficacy must:

      • Correlate with tumor size/volume
        • The actual number of tumor cells in the body → tumor load
      • Short in half-life circulation
      • Be highly specific and sensitive for a single type of cancer

      The ideal tumormarker

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC19: Surgical oncology

      Mechanisms of Disease 2 HC19: Surgical oncology

      HC19: Surgical oncology

      Challenges of cancer

      In 2012, there where 14 million new cancer cases, 8 million cancer deaths and 33 million people living with cancer. These numbers will only increase. Even though surgeons aren’t the only doctors involved in cancer therapy, 80% of all solid cancers need surgery.

      Surgical oncology has several challenges which need to be taken into account:

      • Survival/cure
      • Risk of recurrence
      • Morbidity/mortality of treatment
      • Quality of life/functionality
      • Costs

      There is an evolution in surgery from more invasive to less invasive.

      Types of surgery

      There are several types of surgical oncology:

      • Curative
      • Minimal invasive
      • Acute
      • Palliative
      • Prophylactic

      Curative surgery:

      Curative surgery is an intervention with the aim of curing the disease. Several things happen:

      • En-bloc resection: removal of the primary tumor and all the adjacent tissue which can contain micro-metastases
        • E.g. breast amputation + removal of the nearby lymph nodes
      • No-touch technique: as little as possible has to be done with the tumor itself during its ejection → prevents tumor cells from detaching and metastasizing
        • This can be done with vasculature isolation → the vessels are isolated so the tumor cells can’t travel anywhere
      • Clips: mark the site where the original tumor was → the radiation therapist knows where to provide additional therapy
      • Rinsing the surgical field

      Acute surgery:

      There are 3 situations in cancer where acute surgery is necessary:

      • Perforation
      • Obstruction
      • Bleeding

      What kills first, has to be treated first. An example of this is the removal of a tumor which obstructs the colon → the tumor is removed to restore the normal function. Chances of cure in case of acute surgery are much lower than in normal, elective surgery.

      Palliative surgery:

      Palliative surgery is an intervention with the aim of easing the complaints of the patient. The chance of curation is 0%.

      Palliative surgery may be useful to:

      • Prevent obstruction → creation of a bypass
      • Local control → excision

      Complications of palliative surgery may be:

      • Obstruction
      • Fractures
      • Seroma

      Palliative surgery usually isn’t preformed on old and frail patients.

      Prophylactic surgery:

      Prophylactic surgery is preformed in the tissues where the main tumor often metastasizes in order to prevent further spread of the disease.

      Debulking:

      Debulking is the act of decreasing the number of tumor cells, and thereby removing the major part of the tumor load. This is applied in case of ovarian cancer and is usually followed by chemotherapy.

      Resectability:

      Resectability describes what kind of resection needs to be done:

      • R0: radical resection of the tumor with no tumor cells left in the body
        • The best margin
      • R1: macroscopically yes, but microscopically no
        • Microscopical irregularity is visible
      • R2: residual tumor macroscopically in situ
        • During surgery, the surgeons are cutting through the tumor → residue is left behind

      Less is more

      In surgery, less is always more → less invasion leads to less morbidity.

      This principle can

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC20: Radiation oncology

      Mechanisms of Disease 2 HC20: Radiation oncology

      HC20: Radiation oncology

      Radiation oncology

      Radiation oncology is a separate discipline in oncology where ionizing radiation is used to treat cancer. It is not the same as radiology or nuclear medicine:

      • Radiation oncology uses as much radiation as possible, radiology uses as little radiation as possible
      • Nuclear medicine is done with the use of soluble radionuclides, radiation oncology uses linear accelerators and fixed sources

      50% of cancer patients are irradiated. Radiotherapy can be a form of curative or palliative treatment. Together with other treatments, it increases the numbers of long survivors.

      Ionizing radiation

      There are several types of ionizing radiation:

      • α-radiation
        • Low penetrance
          • Can be stopped by a piece of paper → not often used
        • Sometimes used with cell cultures to measure
      • β-radiation
        • Often used
      • γ-radiation
        • Most used
      • H+-radiation (protons)
        • Sometimes used
      • Electrons
        • Sometimes used
        • Low penetrance
          • Ideal for superficial tumors (skin cancers) → doesn’t affect deeper laying tissues

      External beam radiotherapy:

      External beam radiotherapy uses the photons of γ-rays. A linear accelerator can be turned on or off. This is a form of local treatment → there only is radioactivity when the linear accelerator is on. Afterwards, patients won’t be radioactive anymore.

      Mechanism of action

      Radiation oncology has the following mechanism of action:

      1. Radiation causes DNA-strand breaks
        • This can happen due to direct photon-DNA interaction, but mostly due to indirect interaction: photons activate oxygen radicals → cause DNA damage
        • Double strand breaks, single strand breaks and other kinds of DNA damage are being caused
      2. Normal cells recognize and repair most of the damage
      3. Cancer cells have less regenerative capacity and die at cell division

      The aim is to kill the tumor. The side effects of the therapy are determined by the slow down of cell division in surrounding healthy tissues. This causes both acute and late cell side effects.

      Dose severity

      A higher dose does not always give more tumor control → the likelihood of tumor control isn’t a linear line but is S-shaped. The higher the dose, the higher the toxicity to both tumor and normal cells. The aim of radiotherapy is to kill the tumor without normal cell damage.

      Therapeutic window:

      The therapeutic window or safety window refers to a range of doses which optimize between efficacy and toxicity, achieving the greatest therapeutic benefit without resulting in unacceptable side-effects or toxicity. It is the space between the tumor control curve and toxicity curve.

      There are ways to increase the therapeutic window:

      • Make the tumor more sensitive to radiotherapy
        • The tumor responds much faster than the normal healthy cells
        • The therapeutic window becomes bigger → more tumor control with the same amount of radiation
      • Make the healthy tissue more resistant to radiation
        • A bigger therapeutic index

      Increasing the therapeutic window enhances the sensitivity.

      Side effects

      Radiation therapy has early,

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC21: Medical oncology

      Mechanisms of Disease 2 HC21: Medical oncology

      HC21: Medical oncology

      Oncology

      An oncologist is a doctor who treats cancer and provides medical care for a person diagnosed with cancer. The field of oncology has 3 major areas:

      • Surgery
      • Radiation
      • Medical

      A medical oncologist treats cancer using hormonal therapy, chemotherapy or other medications such as targeted therapy or immunotherapy.

      Breast cancer treatment

      Drug treatments can attack all the cancer cells throughout the body. Most breast cancer cells metastasize to the lymph nodes.

      Goals:

      Goals of breast cancer treatment are:

      • Regression of metastases
      • Improvement in symptoms and quality of life
      • Improvement in survival time
        • There isn’t a cure for metastatic breast cancer yet
      • Balance toxicity of treatment with relief symptoms due to tumors

      Types:

      There are 4 types of therapy for breast cancer:

      • Endocrine therapy
      • Chemotherapy
      • Targeted therapy
      • Immunotherapy

      Factors:

      Factors deciding a certain therapy in metastatic breast cancer are:

      • Patient age
      • Menopausal status
        • Levels of estrogen are much higher in premenopausal women than in postmenopausal women
      • General health
      • Tumor estrogen receptor (ER) status (and the less important progesterone receptor (PgR) status
        • Endocrine therapy is the preferred choice for ER+ metastatic breast cancer → has less side effects than chemotherapy
      • Tumor HER-2 status
        • Targeted therapy is the preferred choice
      • Good/poor responses to previous treatments

      Hormonal therapy

      Hormonal therapy is directed towards tumors which are hormonal dependent for their growth:

      • Breast cancer
        • Estrogen receptor positive (ER+)
      • Prostate cancer
        • Testosterone dependent
      • Endometrial carcinoma
        • Progesterone dependent

      Estrogen:

      Estrogen is a steroid which binds to the estrogen receptor on ER+ breast cancer cells → stimulates tumor cell growth. Approximately 60-70% of breast cancers express estrogen and/or a progesterone receptors. The goal of hormonal treatment is to block the stimulating of cancer growth by steroids.

      There are 2 types of hormonal treatment of ER+ breast cancer:

      • Blockade of estrogen action on the cancer cell
        • Tamoxifen: blocks the estrogen receptors on the surface cells → prevents estrogen from entering the cells
          • A selective estrogen receptor modulator
          • A very old, inexpensive drug
          • Relatively little side effects
        • Treatment for pre- and postmenopausal women with ER+ tumors
      • Blockade of estrogen synthesis in the body
        • Removal of both ovaries in premenopausal women or the use of LHRH agonists
          • Removal of the ovaries isn’t very effective in postmenopausal women → estrogen production hardly goes down
        • Aromatase inhibitors: block the enzyme aromatase → block the conversion of steroid precursors to their active form
          • Anastrozole/armidex
          • Only in postmenopausal women
          • Aromatase is an enzyme in fat, the liver, muscles and the brain → blocks conversions of testosterone and androstenedione into estrogens
            • A source of estrogen in postmenopausal women

      Side effects:

      Side effects of hormonal treatment are very hard to see. Doctors may perceive that the treatment is going well and that

      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC22: Chemoradiation

      Mechanisms of Disease 2 HC22: Chemoradiation

      HC22: Chemoradiation

      Enhancement of sensitivity

      The therapeutic window can be increased by:

      • Fractionation of the total dose
      • Decreasing the toxicity → lower the dose to normal tissue
      • Increasing the tumor control → enhancing the sensitivity

      The sensitivity can be enhanced via:

      • Chemoradiotherapy
        • Radiotherapy + chemotherapy
      • Biological agents
        • Cetuximab in head and neck cancer
      • Oxygen to tumor tissue
        • Increases the oxygen which reaches the tumor → high response rate
        • Doesn’t really work
      • Hyperthermia
        • Exposing tumor tissue to high temperatures
        • In patients with cervical cancer and relapses of breast cancer

      Concomitant and sequential

      Chemoradiation is a combination between radiotherapy and chemotherapy. This can be:

      • Concomitant: in the same time period
      • Sequential: behind each other

      Mechanism of action

      If chemoradiation is used, DNA damage to tumor cells is more often fatal than with irradiation alone. Normal cells have a greater variety of escape mechanisms and repair the damage, which causes them to be more able to repair DNA than tumor cells → have a higher tolerance to the therapy. There are different modes of action for radiation and chemotherapy. Chemotherapy is used as a radiosensitizer which enhances the effect of radiotherapy.

      Synergy:

      Chemo- and radiotherapy can be used to create synergy:

      1. Cisplatin adducts to DNA → causes single strand breaks
      2. Radiotherapy will create more breaks which are more difficult to repair
      3. Post-irradiation repair is inhibited

      Chemotherapy inhibits nucleotide metabolism, while radiation is effective in a different phase of the cell. Chemotherapy treats hypoxic cells, which are less radiosensitive for radiotherapy. Chemoradiation is seen as local treatment to increase local control and thereby cure.

      Uses of chemoradiation

      Chemoradiation is only used in curative treatment:

      • As a primary treatment modality
        • Organ saving treatment
        • Curative resection is not possible
      • As adjuvant treatment
        • Increases local control after resection
      • As neoadjuvant treatment
        • Increases local control after resection

      Primary treatment:

      As primary treatment, chemoradiation can be used as an organ saving treatment. This can be useful when curative resection causes a large loss of function and therefore isn’t possible or when the patient isn’t operative anymore. Examples are:

      • Anal cancer
        • When surgery is done, the patient will most likely lose his anus and need a stoma
      • Head and neck tumors
        • E.g. in case of larynx cancer: if a patient loses their voice, the quality of life decreases significantly
      • Cervical carcinoma
      • Vulvar cancer
      • Esophageal cancer
        • Not operable
      • Stage III lung cancer

      Adjuvant treatment:

      Chemoradiation as adjuvant treatment increases the local control after resection, for example in case of stomach cancer. It can prevent recurrence.

      Neoadjuvant treatment:

      Chemoradiation as neoadjuvant treatment:

      • Increases the local control after resection
      • Downstages resectable cancer

      Examples are operable esophageal cancer and advanced stage rectal cancer.

      Side effects

      Chemoradiation has much more acute side effects in contrast to surgery:

      • Acute side
      .....read more
      Access: 
      JoHo members
      Mechanisms of Disease 2 HC23: Normal hematopoiesis

      Mechanisms of Disease 2 HC23: Normal hematopoiesis

      HC23: Normal hematopoiesis

      Blood cells and functions

      Blood composition:

      Blood forms 8% of the total body weight, the other 92% consists of fluids and tissues. Blood is made up for 55% of blood plasma and for 45% of formed elements. Plasma is mostly made up of water, formed elements of red blood cells, white blood cells and platelets.

      Main functions:

      Blood has many functions:

      • Homeostasis: regulation of body temperature by plasma
      • Body supply: oxygen and nutrients supply by erythrocytes and plasma
      • Waste collection: of carbon dioxide and lactic acid by erythrocytes and plasma
      • Defense: anti-microbial and anti-tumor response by leukocytes
      • Coagulation: by thrombocytes

      Erythrocytes:

      Erythrocytes form the largest fraction of blood cells → there are 5 x 1012erythrocytes per liter blood. They are red cells → cause the red color of blood. Erythrocytes can spend up to 120 days in the circulation. They have no nucleus and are fully differentiated → no proliferation takes place. Their function is oxygen transport.

      Erythrocytes contain hemoglobin:

      • Males: 8,5-11,0 mmol/L
      • Females: 7,5-10,0 mmol/L

      Leukocytes:

      The normal leukocyte count in blood is 4-10 x 109 per liter. Leukocytes can differentiate into:

      • Granulocytes: have a life span of 1-2 days
        • Neutrophilic granulocytes
          • 1,5-7,5 x 109/L
          • Defense against encapsulated bacteria
        • Eosinophilic granulocytes
          • <0,5 x 109/L
          • Mediate allergic responses and defense against parasites
        • Basophilic granulocytes
          • <0,2 x 109/L
          • Mediate allergic responses and defense against parasites
      • Lymphocytes: have a life span of days-years
        • B-cells
          • Antigen-specific immune defense
            • Antibody production
            • Cytokine secretion
        • T-cells: CD4 T-cells and CD8 T-cells
          • Antigen-specific immune defense
            • Cellular cytotoxicity
            • Cytokine secretion
        • NK-cells
          • Antigen-independent defense
            • Tumor surveillance
          • Cytotoxic capacity
      • Monocytes: have a life span of 12 hours
        • Phagocytosis
          • Bacteria
          • Cell debris
        • Secretion of cytokines
          • TNF
          • IL-1
        • Antigen processing and presentation to T-lymphocytes
        • Macrophages are monocytes that reside in the tissue

      Thrombocytes:

      Thrombocytes are present in the blood in a concentration of 150-400 x 109/L. They cause coagulation in case of vessel damage:

      1. Injury to vessel lining triggers the release of clotting factors called von Willebrand factors
      2. Thrombocytes bind to the von Willebrand Factors
      3. The coagulation cascade is activated
      4. Fibrin strands adhere to the plug to form an insoluble clot
      5. Blood clots are made and the vessel wall is repaired again

      Thrombocytes have a lifespan of 8-10 days.

      Dally production:

      Elements are in constant turnover and have different lifespans:

      • Erythrocytes: 150 x 106 per minute
      • Granulocytes: 50 x 106 per minute
      • Thrombocytes: 150 x 106 per minute 

      Production must react to rapid changes in the environment to ensure homeostasis. Only mature elements gain access to the circulation. In case of infection or bleeding, production can increase 3-8 folds.

      Stem cells

      Stem cells are capable of self-renewal, the ability to go through numerous cycles of cell division while maintaining an

      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC24: Diagnostics in hematology

      Mechanisms of Disease 2 HC24: Diagnostics in hematology

      HC24: Diagnostics in hematology

      Case

      A 40-year-old man visits the GP with complaints of fatigue and pallor. The GP thinks the most likely clinical problem is anemia.

      Automatic hematology analyzer

      The GP requests a complete blood count (CBC) → a differential count of Hb, leukocytes, and thrombocytes. Blood goes through an automatic hematology analyzer, a laser which gets scattered when it encounters a blood cell:

      • Forward scatter/low angle: shows the volume of the cell
      • Side scatters/high angle: shows the complexity of the cell (granules, MPC, etc)

      The machine is able to identify the cells based on size and complexity. The results are shown in a diagram, which makes it possible to determine how many and what kind of cells there are.

      Results

      Results of the CBC show:

      • Hb: 4,3 mmol/L → too low
      • Leukocytes: 40 x 109/L → too high
      • Platelets: 70 x 109/L → too low

      Acute leukemia is suspected. In leukemia, there are too many leukocytes compared to other cells. The diversity also is less.

      Microscopic leukocyte differentiation

      Microscopic leukocyte differentiation consists of a blood smear stained with May Grunwald Giemsa. In case of acute leukemia, differentiation of leukocytes is lost → many strange looking leukocytes which all look the same are present.

      Levels of diagnostics

      There are several levels of diagnostics which make it possible to examine different structures:

      • Resections/biopsies: tissue
      • Cytology: cells
      • Molecular pathology: molecules

      Bone marrow examination

      The bone marrow is the principle site of hematopoiesis. Examination of the bone marrow provides additional information for diagnostic clues seen in the peripheral blood:

      • Increased or decreased leukocytes
      • Increased or decreased Hb
      • Increased or decreased platelets
      • Presence of abnormal or immature cells

      Diagnostic tests:

      Several diagnostic tests on the bone marrow tissue can be done:

      • Standard histology: on tissue level
        • H&E: hematoxylin (nucleus) and eosin (cytoplasm)
        • Histochemical stainings (PAS, alcian blue, etc.)
      • Immunohistochemistry: on tissue/cell level
      • Molecular assays: on molecular level

      Bone marrow aspirates are liquid components of bone marrow. On bone marrow aspirates, tests can be done as well:

      • Hematomorphology
        • Distinguishes cell types based on morphological criteria
          • Blasts
          • Mature and immature cells
            • Myeloid
            • Erythroid
            • Megakaryocytic
          • Lymphocytes
          • Plasma cells
          • Macrophages
          • Mast cells
        • Count: 2 x 500 cells
        • Sensitivity: 1%
          • 1 abnormal cell in background of 100 normal cells
        • Expertise, experience and pattern recognition is necessary
        • Acute leukemia: there is way less diversity → everything looks the same
      • Immunophenotyping (cytology)
        • Distinguishes cell types based on presence or absence of proteins or antigens
          • Blasts
          • Mature and immature cells
            • Myeloid
            • Erythroid
            • Megakaryocytic
          • Lymphocytes
          • Plasma cells
          • Macrophages
          • Mast cells
        • Count: 100.000-1.000.000 cells
        • Sensitivity: 0,0001-0,001%
        • Expertise, experience and pattern recognition is necessary
        • Antigens called clusters of differentiation markers (CD markers) are used to identify cells → specific combinations of CD markers on the cell surface are used to identify different cells
          • CD markers are attached to fluorescent
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC25: Myeloid malignancies

      Mechanisms of Disease 2 HC25: Myeloid malignancies

      HC25: Myeloid malignancies

      Myeloid neoplasia

      Myeloid malignancies are divided into 3 categories:

      • Disorders of proliferation
        • Chronic myeloid leukemia
        • Myeloproliferative disorders
      • Disorders of differentiation
        • Myelodysplastic syndromes
      • Disorders of differentiation and proliferation
        • Acute myeloid leukemia

      Chronic myeloid leukemia

      Chronic myeloid leukemia is a disorder of proliferation → it is a stem cell disease.=

      Case:

      A 48-year-old male patient suffers from fatigue, but has no other complaints. Physical examination shows pallor and an enlarged spleen.

      Laboratory tests:

      A laboratory test shows:

      • Hb: 8,0 mmol/L → too low
        • MCV (mean corpuscular volume): 96 fL
      • Leukocytes: 113 x 10E9/L → too high
      • Platelets: 154 x 10E9/L → normal

      Leukocyte differentiation shows:

      • 1 promyelocytes
      • 10 myelocytes
      • 8 metamyelocytes
      • 8 band forms
      • 53 segmented neutrophils

      There shouldn’t be any promyelocytes, myelocytes and metamyelocytes present in the blood. The only cells that should be present are mature blood cells.

      Diagnosis:

      The diagnosis is Philadelphia chromosome positive chronic myeloid leukemia (CML). The driver mutation of CML is BCR/ABL, caused by a translocation of chromosome 9 and 22. This mutation causes cells to have a proliferative advantage. There is no mutation in differentiation genes → differentiation in mature stages is normal. Therefore, neutrophils are segmented as usual.

      Mechanism:

      Chromosome 9 contains the ABL gene, chromosome 22 contains the BCR gene. The mutation takes place as follows:

      1. Both chromosomes break
      2. The ABL gene attaches to the BCR gene on chromosome 22 → the Philadelphia chromosome is created
      3. A constantly active tyrosine kinase is made
      4. Multiple substrates are phosphorylated → multiple ways of absent regulation are made
      5. Chronic myeloid leukemia arises

      It is unknown why the translocation of chromosome 9 and 22 occurs. There is no relation with exposition to chemicals.

      Epidemiology:

      Chronic myeloid leukemia has a slowly progressive course and is a member of the myeloproliferative diseases:

      • Forms 14% of all adult leukemias
      • Incidence: 1-1,5/100.000
      • Male : female = 2:1
      • Median age: 53 years
        • There is an increase in younger patients

      Symptoms:

      Symptoms of chronic myeloid leukemia are:

      • Fatigue and weight loss
      • 40% asymptomatic → disease is detected by chance
      • 50% splenomegaly (enlarged spleen)
      • 20% hepatomegaly (enlarged liver)
      • 50% anemia
      • 20% thrombocytosis

      Target therapy:

      Due to the known mutated shape of BCR/ABL tyrosine kinase, there is a specific inhibitor which binds to the ADP spot of tyrosine kinase → cannot bind to ATP. This causes the signaling pathway to halt. Normally, ATP activates a signaling cascade causing absent regulation.

      Course of the disease:

      Chronic myeloid leukemia has 3 phases:

      1. Chronic phases
      2. Accelerated phase
        • >15% malignant blasts and promyelocytes
        • >20% basophils
        • <100 platelets
      3. Additional cytogenic abnormalities are present
      4. Blast phase: looks like acute leukemia
        • >20% blasts in the blood and bone marrow

      Over time, the normal cells in the bone marrow are outcompeted by the mutated cells. In the later stages of the disease, the mutated

      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC26: Malignant lymphomas

      Mechanisms of Disease 2 HC26: Malignant lymphomas

      HC26: Malignant lymphomas

      Principles of lymphoid differentiation

      There are 2 types of malignant lymphomas:

      • T-cell lymphoma/NK-cell lymphoma
      • B-cell lymphoma

      Pluripotent hematopoietic stem cells (HSC) differentiate into:

      • Common lymphoid precursor cells (CLP) → differentiate into:
        • T-precursor cells → naïve T-cells → T-memory cells
        • B-precursor cells → B-memory cells or plasma cells
        • NK-cells
      • Common myeloid precursor cells (CMP)

      By morphology alone, CLPs cannot be distinguished from CMPs → additional tests are necessary. A plasma cell is recognizable thanks to its big Golgi system. This is the only type of cell which can be distinguished by cell morphology alone. The difference between T-cells and B-cells can be made by looking at cell type specific antigens which are present on the cell:

      • T-cells: CD3
      • B-cells: CD19, CD20, CD79
      • NK-cells: CD16/56

      Neoplasia:

      The primary purpose of lymphocytes is to recognize an antigen receptor. Precursor cells, like blast cells, do not have antigen receptor (AgR) expression yet → they are AgR negative. Both AgR positive and negative cells can cause neoplasia:

      • Precursor neoplasia: caused by AgR negative cells
        • Have a blast-like morphology
      • Mature neoplasia: caused by mature (AgR positive) cells
        • Have a lymphocyte/plasma cell morphology

      If mature cell receptors are destroyed, the cell goes into programmed apoptosis. This doesn’t happen in case of plasma cells, because they don’t have a receptor.

      Physiological B-cell differentiation

      There aren’t enough genes to code for the different type of receptors present in the adaptive immune system. This is solved by VDJ (IgH) and VJ (IgL) recombination:

      1. A B-cell precursor starts to express a receptor to become a B-cell
      2. The B-cell precursor starts to cut out parts of DNA to form a functional gene → multiple V and J sequences are put together
        • 18 different combinations can be made
      3. More differentiation is possible by nibbling away some nucleotides on the open ends of DNA and adding random nucleotides
        • This happens by chance
      4. A completely new functional gene with a completely unique receptor is made

      Mechanism of B-cell lymphomas

      Lymphomas are frequently caused by activation of proto-oncogenes in the course of antigen receptor formation:

      • Chromosomal translocations
        • VDJ recombination
        • Class switch recombination
      • Point mutations
        • Somatic hypermutation

      This results in resistance to apoptosis and activation of signaling cascades.

      B-cell maturation is a dangerous process because it contains double strand DNA breaks → risk of getting mutations. For instance, this can happen when VDJ-recombination activates a proto-oncogene:

      1. During recombination an oncogene is translocated to an immunoglobulin heavy chain locus on the genome
      2. The oncogene becomes irreversibly active
      3. The B-cells containing oncogenes mature
      4. In a lymph node, the mutated B-cell is exposed to an antigen for which its receptor is sensitive
      5. The germinal center response takes place → the mutated B-cell develops from centroblast to centrocyte
        • Somatic hypermutation and class switch recombination takes place
      6. The mutated B-cells have
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC27+28: Allogenic stem cell transplantation and donor lymphocyte infusion I&II

      Mechanisms of Disease 2 HC27+28: Allogenic stem cell transplantation and donor lymphocyte infusion I&II

      HC27+28: Allogenic stem cell transplantation and donor lymphocyte infusion I&II

      What is hematopoietic stem cell transplantation?

      Hematopoietic stem cell transplantation (SCT) is performed because:

      • It is a rescue procedure after a high dose of chemotherapy or irradiation to ensure hematopoiesis
      • It is a way to reset the immune system
      • It replaces non- or dysfunctional bone marrow
      • It is an allogeneic cellular immune therapy for malignant disorders

      Disorders treated with autologous SCT are lymphoma and multiple myelomas. SCT is a form of cellular immunotherapy.

      Harvesting a stem cell graft

      A stem cell graft is harvested as follows:

      1. Filgrastim (neupogen) is injected subcutaneously into the patient
      2. G-CSF receptors are activated → cells migrate out of the bone marrow into the circulation
      3. A certain machine removes the cells which the patient doesn’t need, and returns the other cells back to the patient

      Autologous stem cell transplantation

      Autologous SCT is performed as follows:

      1. The stem cell graft is harvested from the patient
      2. Cells are cryopreserved in liquid nitrogen
      3. The patient is treated with high dose chemotherapy → results in ablation of the stem cells from the bone marrow
      4. The stem cell product is reinfused
        • All stem cell harvests contain a mixture of leukocytes: stem cells, T-cells, B-cells, NK-cells, monocytes and granulocytes

      The actual treatment is the chemotherapy → stem cell transplantation only is a rescue procedure.

      Allogeneic hematopoietic SCT

      The difference between allogeneic and autologous SCT is that in allogeneic SCT the graft of bone marrow cells is taken out of a healthy donor instead of the patient.

      Procedure:

      Allogeneic SCT is performed as follows:

      1. A stem cell graft is harvested from the donor
        • Using G-CSF treatment
      2. The patient’s immune system and leukemia cells are destroyed
        • Immune ablation of the patient is necessary to allow acceptance of donor cells → a stem cell graft is highly immunogenic
      3. Chemotherapy is used to make space for donor stem cells in the bone marrow
      4. Sometimes the stem cell product is modified → is optional
      5. A donor stem cell collection is infused into the patient, who doesn’t have an immune system
        • This can include donor T-lymphocytes
      6. The immune system is rebuilt with donor cells → the patient has a new hematopoietic system

      Appliances:

      Allogeneic SCT is used to treat:

      • Genetic disorders or failure of hematopoiesis or lymphopoiesis
      • Acquired stem cell failure syndromes
      • Hematological malignancies
        • Autologous SCT cannot be performed because autologous stem cells harbor the mutation/abnormality of the original stem cell disease

      Rejection:

      Allogeneic hematopoietic SCT is a very complex procedure because it messes with 2 different immune systems:

      • T-cells from the patient can reject the stem cells → host versus graft disease
      • T-cells in the graft coming from the donor can react with various cells from the patient → graft versus host disease

      There are 2 ways to prevent graft rejection:

      • Suppression of T-cells from the patient via
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC29: HLA & minor histocompatibility antigens

      Mechanisms of Disease 2 HC29: HLA & minor histocompatibility antigens

      HC29: HLA & minor histocompatibility antigens

      Allogeneic SCT

      There are 2 ways of doing an allogeneic SCT:

      • Allogeneic SCT with T-cells present in the cell graft: transplantation of an immune system of a healthy donor with the aim to induce a strong T-cell response against the cancer cells of the patient
        • Mature donor T-cells in the stem cell transplant mediate GVL and GVHD
        • Systemic immunosuppression is required to suppress GVHD
        • Systemic immunosuppression is gradually decreased
      • Allogeneic SCT without T-cells present in the cell graft
        • No systemic immunosuppression is required
        • Consists of 2 steps
          • T-cell depletion → no systemic immunosuppression is required
          • DLI is necessary: 3-6 months after allogeneic SCT to induce GVL
            • The patient is in better condition
            • Professional antigen-presenting cells of donor origin
            • Less pathogens
            • Less inflammatory cytokines
        • The T-cell response is weaker → better balance between GVL and GVHD
          • These patients do better as a group → lesser GVHD reaction
            • The body has time to heal damaged tissue from chemotherapy
            • Recipient dendritic cells can be replaced by donor dendritic cells

      T-cells in infections

      HLA:

      HLA molecules are located on chromosome 6. Per chromosome, there are 6 genes which play a role in HLA. Because humans have 2 chromosomes, there are 12 genes in total which play a role. There are 2 types of HLA molecules:

      • HLA class I: present intracellular antigens
        • HLA-A
        • HLA-B
        • HLA-C
      • HLA class II: present endocytosed antigens
        • HLA-DR
        • HLA-DQ
        • HLA-DP

      The HLA groups are located in the peptide binding groove. Only dendritic cells, macrophages and B-cells are capable of HLA-II expression. HLA is highly polymorphic → there are many variants and every different allele has its own name.

      Negative selection:

      Due to negative selection in the thymus of T-cells which have high affinity for HLA self-complexes, there are no T-cells for processing peptides derived from cellular proteins, otherwise autoimmune reactions would be induced.

      T-cells after allogeneic SCT:

      Donor T-cells recognize foreign peptide-HLA complexes (allo-antigens). When selecting a matching donor, not all 12 but only 10 genes are taken into account → HLA-DP is usually not taken into account:

      • When an unrelated donor (URD) is selected, there usually is a 10/10 match but an HLA-DP mismatch
      • When a family donor (sibling donor) is selected, there usually is a 12/12 match with HLA-DP also matching

      Therefore, after an unrelated allogeneic SCT, there can be T-cells present in the donor graft which are directed against peptides in mismatched HLA-DP → immune reaction. HLA molecules are major histocompatibility antigens. In case of shared HLA molecules, immune responses against minor histocompatibility antigens can occur.

      Minor histocompatibility antigens

      A minor histocompatibility antigen (MiHA) can be:

      • A polymorphic peptide that differs in amino acid composition between patient and donor
      • A polymorphic peptide that is presented on patient cells by HLA surface molecules
        • HLA is matched between patient and donor
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC30: Changes in patients’ experiences

      Mechanisms of Disease 2 HC30: Changes in patients’ experiences

      HC30: Changes in patients’ experiences

      Patients versus medical professionals

      While patients experience cancer treatment as a unique life-changing series of events, to medical professionals it constitutes routine work practice.

      Joanna Baines

      Joanna Baines wrote an article containing 3 stories of 3 generations of breast cancer:

      • Grandmother
        • Stayed at home after diagnosis, died at the age of 56
        • For her, cancer was an experience of silence
      • Mother
        • Was diagnosed with breast cancer at 35
        • Treatment
          • Radical mastectomy
            • Removal of the entire breast, nipple, lymph nodes, vessels and muscles → severe and mutilating operation
            • Became less and less popular throughout the 1970s
          • Ovarian ablation throughout radiotherapy
          • 5 year follow up of remaining breast and glands
        • Hardly received any information about her condition → saw cancer as an event
      • Joanna Baines
        • Diagnosed with breast cancer at 32 years old
        • Received several supplementary tests
          • Abdomen
          • Liver
          • Bones
        • Treatment
          • Removal of the lump and lymph nodes in the armpit
          • Chemotherapy (5 months)
          • Radiotherapy
          • Tamoxifen (5 years)
          • Herceptin
        • She was part of an RCT (randomized control trial)
        • Saw cancer as a part of her life

      Timeline

      A timeline of emerging cancer therapies has been created:

      • 1850-1920: radical surgery
      • 1900-1950: radiation
        • Major alternative to surgery before chemotherapy
        • High energy radiation equipment
          • Huge machine surrounding the patient
          • Emerged in the early 20th century
      • 1950-1970: chemotherapy
        • Emerged after the WWII
        • 3 types
          • Nitrogen mustards
            • Discovered during WWII
            • Don’t produce lasting remission
          • Hormones
            • Increased use in the 1940s-1950s
            • Now regarded as palliative
            • Tamoxifen became available in the 1970s-1980s
          • Antimetabolites
            • Sydney Farber diagnosed ALL in a 2-year-old and injected him with aminopterin (an antifolic) → worked very well
        • Didn’t become the cure that everyone had hoped → only prolonged the lives for several months
        • Issues
          • Drug resistance
          • Death due to new complications
          • Survival isn’t worthwhile
        • Many experiments where done in the 1960s
      • 1970-1990: combination therapy
        • Dr. Pinkel made a breakthrough by combining high dose chemotherapy and radiation
          • More toxic chemicals
          • Radiotherapy in the brain and chemotherapy in the spinal fluid
          • Double doses in case of no success
          • “Total therapy” → a total hell for patients
        • Even though there was a huge therapeutic optimism, there were several problems
          • Toxicity, response, life impact and trauma, complications, drug resistance, etc.
          • Ethical dilemmas in research of cancer treatment
        • 1978: cisplatin was introduced
          • Highly effective
          • Many toxic effects → kidney failure later on
      Access: 
      Public
      Mechanisms of Disease 2 HC31: Targeted therapy and hematological malignancies

      Mechanisms of Disease 2 HC31: Targeted therapy and hematological malignancies

      HC31: Targeted therapy and hematological malignancies

      Monoclonal antibodies

      Tumor specific monoclonal antibodies:

      CD20 is an IgG isotype expressed antigen on normal and malignant B-cells. It is used for treatment of many B-cell lymphomas → by targeting CD20 antigens, both malignant and normal B-cells are depleted.

      Rituximab:

      Chemotherapy combined with CHOP monoclonal antibodies leads to a significant increase of survival rates. Rituximab, a monoclonal antibody against CD20, leads to:

      • Complement system activation
        • A MAC is formed
      • NK-cell recruitment
        • Receptors bind to the humanized tail of rituximab
      • Apoptosis
        • Macrophages are attracted

      Mechanisms of action:

      Monoclonal antibodies can have different mechanisms of action:

      • Complement dependent cytotoxicity (CDC)
        1. The complement system is activated
        2. Hollow pipes are made in the cell surface
        3. Water enters the cell
        4. The cell explodes
      • Antibody dependent cellular cytotoxicity (ADCC)
        1. A B-lymphocyte plasma cell secretes antibodies which bind to an antigen on the pathogen
        2. The pathogen is phagocytosed by macrophages, destroyed by NK-cells or lysed via the complement system
      • Radio immunotherapy
        1. A radioactive particle such as yttrium is coupled to an IgG monoclonal antibody such as CD20 → can get close to the tumor cells
          • A form of local treatment
        2. Yttrium gives a small dose of β- or γ-radiation
        3. The tumor is directly targeted
      • Antibody drug conjugate
        1. A drug is bound to a monoclonal antibody such as CD30 → an ADC-CD30 complex is created
          • CD30 is expressed in Hodgkin lymphomas and anaplastic large cell lymphomas
        2. The ADC-CD30 complex travels to the lysosome
        3. MMAE is released in the cell → disrupts the microtubule network
          • Physical cell division is prevented by binding on a microtubule during cell division
        4. The G2-/M-phase is arrested → the cell cycle stops → apoptosis

      Bispecific antibodies

      Bispecific antibodies are a combination of 2 different antibodies to target B-cells with CD8 T-cells. A bispecific T-cell engager (BiTE) causes proliferation of T-cells via the following mechanism:

      1. 2 antibodies are coupled to create a bispecific monoclonal antibody with binatunumab → an antibody against CD3 antibody is coupled with an antibody against CD19
        • Anti-CD3 antibodies target T-cells
        • Anti-CD19 antibodies target B-cells
      2. The T-cell becomes activated and starts its immune function → cytokines are made
        • A T-cell can kill more cells than 1 B-cell
      3. The T-cell attacks malignant B-cells → perforins enter the B-cells and kill them

      CAR T-cells

      A T-cell has a T-cell receptor, which can recognize a peptide presented in an HLA molecule. To activate a T-cell receptor, additional costimulatory signals such as CD28 are necessary as well. If a T-cell receptor is activated, the T-cell proliferates and kills the target cell. A Chimeric antigen receptor (CAR) T-cell uses the intracellular part of a TCR and the extracellular part of a monoclonal antibody:

      1. T-cells are extracted through a leukapheresis procedure
      2. T-cells are expanded if necessary
        • Usually
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC32+33: Primary hemostasis

      Mechanisms of Disease 2 HC32+33: Primary hemostasis

      HC32+33: Primary hemostasis

      Cancer and thrombosis

      Thrombophlebtis is thrombosis due to infected vessels. Thrombotic events are the second leading cause of death in cancer patients, after death from cancer itself. The Trousseau syndrome describes the presence of thrombosis in relationship to cancer. Fibrin clots may be induced by cancerous processes:

      • Cancer has an influence on coagulation → influences thrombosis
      • Coagulation may have an influence on how the cancer will develop
      • Thrombosis also has an effect on developing cancer.

      Risk:

      Patients with cancer have an increased risk for developing venous thrombosis:

      • Odds ratio: 7
      • Incidence: 24/1000/year
      • Risk and time since diagnosis of cancer
        • 0-3 months: odds ratio of 54
        • 3-12 months: odds ratio of 14
        • 12-36 months: odds ratio of 4

      The Khorana risk score predicts the risk of cancer associated thrombosis. This risk differs per cancer type and patient:

      • Site of cancer
        • Very high risk → 2
          • Stomach cancer
          • Pancreas cancer
        • High risk → 1
          • Lung cancer
          • Lymphomas
          • Gynaecological cancer
          • Bladder cancer
          • Testicular cancer
      • Prechemotherapy platelet count >350 x 109/L → 1
      • Hemoglobin level <10 g/dL or use of red cell growth factors → 1
      • Prechemotherapy leukocyte count >11 x 109/L → 1
      • Body mass index >35 kg/m2→ 1

      These scores can be added to predict the risk:

      • >3 → high risk
      • 1 or 2 → intermediate risk
      • 0 → low risk

      Patients with thrombosis also have a risk of developing cancer:

      • Idiopathic (unprovoked) venous thrombosis: 10% of patients develop cancer in the next 3 years
        • Unprovoked: thrombosis with no other clear cause
      • Provoked venous thrombosis: 1,6% of patients develop cancer in the next 3 years
      • Single episode of superficial thrombosis: no clear association with cancer
        • If it returns, there is a connection

      Mortality:

      Cancer and thrombosis are a very bad combination. Extensive screening can identify occult tumors, but this does not improve the survival and can lead to false positive cases. Therefore, currently just a general screening is done.

      Treatment of venous thrombosis:

      In cancer patients, anticoagulation with low molecular weight heparin (LMWH) is more effective than vitamin K antagonists in prevention of recurrent venous thrombosis.

      Older thrombosis studies also show a potential benefit of anticoagulant treatment. LMWH shows an improved 1-year survival only in patients with localized cancer. This is only a small advantage which isn’t proven. Even though there is a clear correlation, the conclusion is that there is no benefit of LMWH on cancer.

      Mechanism of hemostasis

      There are many factors which interfere with the coagulation system. As soon as the endothelium layer is broken or compromised, the coagulation begins:

      1. First pathway: after the endothelium layer is compromised, vasoconstriction leads to a reduced blood flow
      2. Second pathway: the blood gets into contact with subendothelial collagen → platelet adhesion and aggregation
        • A platelet plug is made
        • Serotonin and TxA2 are secreted → enhance vasoconstriction
      3. Third
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC34+35: Secondary hemostasis I&II

      Mechanisms of Disease 2 HC34+35: Secondary hemostasis I&II

      HC34+35: Secondary hemostasis I&II

      Secondary hemostasis

      Blood flows as a fluid through the blood vessels to all the organ systems. Upon injury, blood vessel loss has to be prevented. This is done via blood coagulation. Under normal conditions, blood coagulation is a tightly regulated process:

      • Rapid formation of a clot upon injury
      • Limited to the site of injury

      Secondary hemostasis is the conversion of soluble fibrinogen to insoluble fibrin by thrombin (IIa):

      • Strengthens the platelet plug
      • Induces adherence and activation of cells involved in vascular repair
        • Fibroblasts
        • Smooth muscle cells

      Thrombin plays a key role in the conversion of fibrinogen to fibrin. It sometimes also is called coagulation factor IIa.

      Fibrinogen

      Fibrinogen is also known as factor I and is very abundant in the plasma → normal levels are 1,5-3 gram/L. It consists of 2 symmetrical half-molecules, each consisting of 3 different polypeptide chains:

      • Aα: linked together in the E-domain → the central parts
      • Bβ: in the D-domain → the outer parts
      • γ: in the D-domain → the outer parts

      Formation of fibrin:

      Fibrinogen is formed into fibrin as follows:

      1. Thrombin cleaves away fibrinopeptides A and B → fibrin monomers are formed
        • Fibrin monomers are very sticky
      2. 2 fibrin monomers stick together → dimers are formed
        • This is done via H-bonds
      3. Coagulation factor XIIIa crosslinks different dimers → polymers are formed
        • Coagulation FXIIIa = transglutaminase
          • The enzyme factor XIII is converted into FXIIIa by thrombin (IIa)
        • The crosslinks are formed between the D-domains of the fibrin
        • These are covalent bonds

      Thrombin

      Thrombin is generated via the coagulation cascade. The coagulation cascade is a sequence of proteolytic reactions, which take place in parts of the surface of activated proteins. In each step, a pro-enzyme is converted to an enzyme, which activates the next pro-enzyme. These are slow reactions which can be sped up by co-factors → a kind of catalysts.

      The coagulation cascade consists of 2 pathways:

      • Extrinsic pathway: activation upon contact with tissue factor (TF)
        • TF was previously named coagulation factor III
        • The initiator is located outside the plasma
      • Intrinsic pathway: activation upon contact with surfaces foreign to our body
        • E.g. glass → contact activation
        • Everything is located inside the plasma

      The common pathway consists of the coagulation factors which play a part in both intrinsic and extrinsic pathways:

      • FX
      • FV
      • FII

      Extrinsic pathway:

      There are several very important factors in the extrinsic pathway:

      1. Tissue factor, factor VII
      2. Factor X, factor V
      3. Factor II: prothrombin

      Tissue factor (TF) is a transmembrane glycoprotein and a non-enzymatic co-factor of FVIIa. TF is produced by fibroblasts and smooth muscle cells → is expressed on a "hemostatic envelope” around blood vessels. TF normally isn’t present in the blood compartment, but during pathological conditions it can be expressed:

      • Inflammation: expression on endothelial cells and monocytes
        • This is
      .....read more
      Access: 
      Public
      Mechanism of Disease 2 HC36: Fibrinolysis and atherothrombosis

      Mechanism of Disease 2 HC36: Fibrinolysis and atherothrombosis

      HC36: Fibrinolysis and atherothrombosis

      Fibrinolysis

      At some point, the platelet plug needs to be removed to return the normal blood flow.

      Fibrinolysis is part of the normal hemostasis. It enables repair of the injured vessel wall and is important in resolving an unwanted clot such as a thrombus.

      Fibrinolysis consists of the conversion of earlier formed fibrin, which is insoluble, into soluble fibrin degradation products. This process is important because it prevents venous thrombosis and atherothrombosis by removing unwanted clots. It is part of normal hemostasis.

      Plasmin:

      Plasmin is a molecule which degrades fibrin → after degradation, only D-dimers of fibrin are left:

      1. Cross-linked fibrin is degraded by fibrin degradation products
        • Plasmin cleaves the E-domains
      2. D-dimers are formed

      A D-dimer is also a biomarker for the fact that thrombosis is present elsewhere.

      Plasmin generation

      Plasmin generation consists of a sequence of proteolytic reactions on the surface of fibrin. There are 2 pathways, dependent of the activator:

      • Pathway 1: tPA dependent plasmin generation
        • Activator: tissue plasminogen activator (tPA)
          • Produced by healthy endothelial cells
        • tPA converts plasminogens to plasmins
          • Fibrin is the co-factor in his reaction → speeds up the reaction
            • tPa is only active when bound to fibrin → advantageous in thrombosis
            • Is the surface where tPA and plasmin react together
      • Pathway 2: uPA dependent plasmin generation
        • Activator: sc-uPA or pro-urokinase
          • Produced by endothelial cells, macrophages and epithelial cells
          • Plasmin converts sc-uPA to tc-uPA
            • Tc-uPA is the active form of sc-uPA
        • tc-uPA converts plasminogen to plasmin → amplification
        • The reaction takes place in both presence and absence of fibrin

      Regulation:

      Plasmin generation is regulated by:

      • Presence of tPA and uPA
      • Inhibitors of tPA, uPA and plasmin
        • Plasminogen activator inhibitor 1 (PAI-1)
          • Produced by endothelium, platelets and monocytes
          • PAI-1 inhibits tPA and uPA highly efficiently
        • α2-antiplasmin (α2AP)
          • Produced by the liver
          • Limits the diffusion of plasmin into the circulation
          • A fast and highly efficient inhibitor of plasmin
      • Fibrin inhibitors
        • Thrombin activatable fibrinolysis inhibitor (TAFI)
          1. TAFI is a plasma pro-carboxypeptidase which circulates in the blood in an inactive form
          2. Thrombin activates TAFI to TAFIa
          3. TAFIa removes lysins from the fibrin surface to which the tPA and fibrinogen bind → inhibition of the conversion of plasminogen to plasmin
          4. Plasmin generation and fibrin degradation are delayed

      Thrombosis and fibrinolysis

      There is a correlation between thrombosis and fibrinolysis:

      • Venous thrombosis and fibrinolysis
        • Cause: thrombi are formed in large veins under low flow (stasis)
        • Therapy: targeting coagulation
          • Heparin antagonists
          • Vitamin K antagonists
          • DOACs
        • Primary aim: preventing occurrence of a pulmonary embolism (PE)
        • The thrombus is removed by endogenous fibrinolysis
      • Arterial thrombosis and fibrinolysis
        • A diseased arterial wall triggers thrombosis
          • This may obstruct the arterial flow
          • Atherosclerotic plaques induce the activation of hemostasis
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC37: Cancer, coagulation and thrombosis

      Mechanisms of Disease 2 HC37: Cancer, coagulation and thrombosis

      HC37: Cancer, coagulation and thrombosis

      The link between cancer and thrombosis

      Cancer leads to coagulation, which leads to thrombosis. Coagulation itself also leads to cancer, and thrombosis may as well. The link between cancer and thrombosis was established almost 200 years ago by Jean-Baptiste Bouillaud and Armand Trousseau. Trousseau diagnosed himself with thrombosis and predicted he suffered of cancer. Months later, he indeed died of pancreatic cancer.

      Dual problem:

      Cancer and thrombosis are a dual clinical problem. Out of 7 million cancer-associated deaths, 1 million are attributed to thrombotic complications. Patients with cancer and thrombosis have an extremely bad prognosis. The second main cause of death in cancer patients is deep venous thrombosis/pulmonary embolism (VTE):

      • 25% of all first VTE events are cancer-related
      • The presence of malignancy results in a 7x increased risk of VTE
        • 0-3 months after cancer diagnosis: 54x increased risk
        • 3-12 months after cancer diagnosis: 14x increased risk
        • 12-36 months after cancer diagnosis: 4x increased risk
      • 8% of all cancer patients experience VTE

      The risk of developing VTE in case of cancer can be divided into 2 groups:

      • Clinical risk factors
      • Biological risk factors

      Clinical risk factors

      Stage:

      Not all cancer types confer the same risk for VTE. Patients with pancreas, lung and/or brain cancer have the highest incidence rate of VTE. The more aggressive the tumor, the higher the risk. The risk of VTE reflects the stage of the disease → in remote cancers, the risk of VTE is highest.

      Therapy:

      Certain cancer treatments can increase the risk of VTE as well:

      • Surgery: 2x increased risk of VTE in cancer patients
      • Chemotherapy: the risk varies
      • New antiangiogenic drugs
      • Prolonged bed rest

      Treatment of venous thrombosis in cancer patients usually consists of low molecular weight heparin (LMWH), which is more effective than vitamin K agonists. LMWH can be prescribed for 6 months. If VTE still is present after this, it is necessary to switch to vitamin K antagonists.

      Biological risk factors

      It is unknown which biological factors cause cancer-associated VTE. Possible causes are:

      • Microparticles
      • Neutrophil extracellular traps (NETs)
      • Coagulation factors
      • Localization of cancer cells to the blood (CTCs)
      • Platelets/leukocytes
      • Compression of blood vessels by the tumor

      Microparticles:

      Microparticles (MPs) are vesicles of 50 nM-1 μM shed from various cells. They are meant for intercellular communication and contain proteins and microRNA. TF+ microparticles (MP-TF) can be shed from:

      • Platelets
      • Leukocytes
      • Endothelium
      • Cancer cells

      MP-TF predicts the chance of VTE → if more MP-TF+ is present, the cumulative incidence of VTE is higher:

      • Healthy volunteers have low TF activity in plasma
      • Cancer patients without VTE have low TF activity in plasma
      • Cancer patients with VTE have high TF activity in plasma → microparticles play a role in VTE

      A hypothesis is that once MP-TF is in the blood, it fuses with platelets, endothelial cells or the ECM. MPs can be measured with:

      • Fluorescence-assisted cell sorting (FACS)
        • Cells flow through a machine
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC38: Bleeding disorders

      Mechanisms of Disease 2 HC38: Bleeding disorders

      HC38: Bleeding disorders

      Causes

      A bleeding disorder can roughly have 3 different causes:

      • Decrease in procoagulant factors
        • Factor VIII
        • Factor IX
        • Factor V
      • Use of anticoagulant medication
        • Vitamin K antagonists
        • Direct oral anticoagulants
      • Disorders such as liver failure, leukemia or immune thrombocytopenia

      Bleeding disorders can be congenital or acquired:

      • Congenital disorders
        • Hemophilia
        • Von Willebrands disease
        • Platelet disorders
        • Deficiencies of other clotting factors
      • Acquired disorders
        • Liver failure
        • Haematological diseases
        • Autoimmune diseases
          • Immune thrombocytopenia
          • Acquired hemophilia
        • Diffuse intravascular coagulation
        • Use of medication
          • Vitamin K antagonists
          • Direct oral anticoagulants
          • Thrombocyte aggregation inhibitors
            • Aspirin
            • Clopidogrel

      Analysis

      Bleeding disorders are analyzed based on:

      • Patient history
        • Congenital versus acquired
        • Type of bleeding
          • Site
            • Mucosal
            • Skin
            • Gums
            • Nosebleeds
            • Menstrual bleeding
            • Joints muscles
          • When it started
            • Spontaneous
            • After trauma
            • After surgery
            • Tooth extractions
        • Other diseases
        • Use of medication
        • Family history
      • Physical examination
        • Signs of bleeding
          • Haematoma
          • Petechiae
            • Red spots under the skin
          • Bleeding in joints or muscles
            • E.g. swollen knees
        • Splenomegaly or lymph node enlargement
          • These are signs of hematological malignancies
        • Skin abnormalities
          • Atypical eczema can be the cause instead of bleeding disorders
      • General laboratory investigations
        • Hb and thrombocyte count
        • Prothrombin time (PT)
        • APTT
        • Fibrinogen
        • Liver and kidney function tests
      • Specific laboratory investigations
        • Bleeding time
        • Platelet function analyzer (PFA)
        • Platelet aggregation tests
        • Measurement of individual coagulation factors
          • FVIII, IX, V, VII, II, X, XI, XII, XIII
        • Measurement of fibrin degradation products
          • D-dimers
        • Inhibitor assays

      Von Willebrands disease

      In case of von Willebrands disease (VWD), there is an abnormality in both primary and secondary hemostasis. The frequency of VWD is <1-1%. It inherits autosomal → men and women are equally affected. There are 3 types of VWD:

      • Type 1: moderate lowering of vWF
      • Type 2: activity of vWF is lower than the antigen level
      • Type 3: severe lowering of vWF
      • Normandy: less binding of factor VIIIc
        • Comparable to mild hemophilia

      Clinical presentation:

      VWD is characterized by the following symptoms:

      • Skin and mucosal bleeding
      • Nosebleeds
      • Menorrhagia
      • Bleeding after trauma and surgery
      • Much less muscle and joint bleeds compared to hemophilia

      In general, the bleeding tendency is much less severe than in case of hemophilia.

      Treatment:

      Treatment of VWD can consist of:

      • DDAVP
        • Releases vWF and factor VIII from Weibel-Palade bodies
        • Effectiveness depends on the type of VWD
          • Ineffective in case of type 2b
          • Testing is necessary
        • Side effects
          • Headache
          • Flushing
          • Hypotension
          • Fluid retention
            • Patients need to drink less water
          • Hyponatriemia
      • Clotting factor concentrates
        • Intermediate purity factor VIII concentrate
        • Contains both vWF and factor VIIIc
        • Plasma derived
        • Recombinant concentrate
      .....read more
      Access: 
      Public
      Mechanisms of Disease 2 HC39: Thrombosis

      Mechanisms of Disease 2 HC39: Thrombosis

      HC39: Thrombosis

      Cause

      Thrombosis is caused by a decrease in anticoagulant factors. Deep vein thrombosis (DVT) has the following symptoms:

      • Red skin
      • Warm
      • Painful calf
      • Swelling
      • Shining skin
      • Pain while walking

      In 40% of cases with suspected DVT, the clinical diagnosis is correct. Ultrasounds of the vessels in the leg are primarily used to diagnose DVT. In normal situations, the vein compresses and becomes hard to see. In case of thrombosis, the vein doesn’t become compressed as much. DVT differs from arterial thrombosis because it occurs at sites of stasis.

      Post thrombotic syndrome

      Post thrombotic syndrome (PTS) is a persistent swollen and painful leg as a result of DVT. Symptoms are:

      • Swollen leg
      • Heavy feeling leg
      • Pain
      • Varicose veins
      • Discoloration of the skin
      • Ulceration

      In post thrombotic syndrome, accumulation of thrombotic platelets causes a leaky valve. Current treatment to prevent the development of post-thrombotic syndrome consists of compression stockings → the venous pressure is increased.

      Pulmonary embolism

      A pulmonary embolism (PE) is an embolization of a venous thrombus into the pulmonary artery. Symptoms are:

      • Breathlessness
      • Pleural pain
      • Hemoptysis
      • Fever
      • Shock

      A very deadly form of pulmonary embolism is a saddle embolism → almost always results in death.

      Imaging:

      Diagnosis of pulmonary embolism requires objective imaging:

      • CT-scans
        • Are primarily used
        • White contrast fluid is injected → gray areas show blood clots
      • Chest X-rays
        • In acute phases of pulmonary embolisms
        • Rules out other diseases mimicking pulmonary embolism
        • After a few days, pulmonary infiltrates may become visible → infarcts
      • Ventilation-perfusion scan: shows radioactivity in the lungs → if there is radioactivity, no blood is present
        • Perfusion: intravenous injection of radioactive technetium macro-aggregated albumin
          • Tc99m-MAA
        • Ventilation: inhalation of gaseous radionuclides in aerosol
          • Xenon-133
      • Pulmonary angiography
        • Was used in the past
        • Contrast fluid is injected into the pulmonary artery with a catheter

      Diagnostic strategies:

      Diagnostic strategies for pulmonary embolism may consist of an algorithm, such as the clinical decision rule:

      • Symptoms of DVT → 3
      • Heart rate >100/min → 1,5
      • Immobilization >3 days or operation in the past 4 weeks → 1,5
      • DVT or PE in medical history → 1,5
      • Hemoptysis → 1
      • Malignancy → 1
      • PE is more likely than any alternative diagnosis → 3

      In case there are less than 4 points, a D-dimer test is done:

      • D-dimers <500 ng/ml → no PE
      • D-dimers >500 ng/ml → possibly PE

      In case there are more than 4 points or D-dimers are >500 ng/ml, a CT or perfusion scan is made:

      • Normal → no treatment
      • PE → treatment
      • Other diagnosis → treatment

      Currently, a more simple way is used → the YEARS algorithm. If 3 YEARS items are present, a D-dimer test needs to be ordered:

      • Clinical signs of DVT
      • Haemoptysis
      • If pulmonary embolism is the most likely diagnosis

      This prevents a lot of unnecessary CT-scans.

      Treatment

      The primary aim

      .....read more
      Access: 
      Public
      Work for WorldSupporter

      Image

      JoHo can really use your help!  Check out the various student jobs here that match your studies, improve your competencies, strengthen your CV and contribute to a more tolerant world

      Working for JoHo as a student in Leyden

      Parttime werken voor JoHo

      Check more of this topic?
      How to use more summaries?


      Online access to all summaries, study notes en practice exams

      Using and finding summaries, study notes en practice exams on JoHo WorldSupporter

      There are several ways to navigate the large amount of summaries, study notes en practice exams on JoHo WorldSupporter.

      1. Starting Pages: for some fields of study and some university curricula editors have created (start) magazines where customised selections of summaries are put together to smoothen navigation. When you have found a magazine of your likings, add that page to your favorites so you can easily go to that starting point directly from your profile during future visits. Below you will find some start magazines per field of study
      2. Use the menu above every page to go to one of the main starting pages
      3. Tags & Taxonomy: gives you insight in the amount of summaries that are tagged by authors on specific subjects. This type of navigation can help find summaries that you could have missed when just using the search tools. Tags are organised per field of study and per study institution. Note: not all content is tagged thoroughly, so when this approach doesn't give the results you were looking for, please check the search tool as back up
      4. Follow authors or (study) organizations: by following individual users, authors and your study organizations you are likely to discover more relevant study materials.
      5. Search tool : 'quick & dirty'- not very elegant but the fastest way to find a specific summary of a book or study assistance with a specific course or subject. The search tool is also available at the bottom of most pages

      Do you want to share your summaries with JoHo WorldSupporter and its visitors?

      Quicklinks to fields of study (main tags and taxonomy terms)

      Field of study

      Access level of this page
      • Public
      • WorldSupporters only
      • JoHo members
      • Private
      Statistics
      1144
      Comments, Compliments & Kudos:

      Add new contribution

      CAPTCHA
      This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
      Image CAPTCHA
      Enter the characters shown in the image.
      Promotions
      vacatures

      JoHo kan jouw hulp goed gebruiken! Check hier de diverse studentenbanen die aansluiten bij je studie, je competenties verbeteren, je cv versterken en een bijdrage leveren aan een tolerantere wereld